31.07.2013 Views

Book 1.indb - Journal

Book 1.indb - Journal

Book 1.indb - Journal

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Scientifi c Tools<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012<br />

CONTENTS<br />

RGUHS <strong>Journal</strong> of Pharmaceutical Sciences<br />

Vol 2, Issue 1, Jan–Mar, 2012<br />

• Knowing the known to understand the unknown: a systematic approach to reviewing<br />

the scientifi c literature 1<br />

Yamuna V. Kuberappa, Arun H.S. Kumar 10.5530/rjps.2012.1.1<br />

Meeting Report<br />

• Pharmaceutical discovery and development 8<br />

(A report on the symposium held in 63rd annual conference of the Indian Pharmaceutical Congress-2011)<br />

Pitchai Balakumar, Gowraganahalli Jagadeesh 10.5530/rjps.2012.1.2<br />

Review Articles<br />

• Nanobiotechnology: An overview of drug discovery, delivery and development 14<br />

Bhupinder Singh Sekhon 10.5530/rjps.2012.1.3<br />

• Nasal drug delivery–a review 24<br />

Twarita Deshpande, Rajashree Masareddy, Archana Patil 10.5530/rjps.2012.1.4<br />

Research Articles<br />

• Design and optimization of levofl oxacin gastroretentive tablets 38<br />

D. Nagendrakumar, S.B. Shirsand, M.S. Para, A.D. Chauhan 10.5530/rjps.2012.1.5<br />

• Study of the binding properties of hydroxypropyl guar and its utilization in the formulation<br />

and evaluation of metoprolol tartarate tablets 45<br />

Swamy N.G.N., Dharmarajan T.S. Paranjothi K.L.K. 10.5530/rjps.2012.1.6<br />

• Phytochemical investigation of root extract of the plant Carissa spinarum 55<br />

Karunakar Hegde, D. Satyanarayana, Arun B. Joshi 10.5530/rjps.2012.1.7<br />

• Synthesis and antimicrobial activity of 4-hydroxy-1-methyl/phenyl-3- (substituted anilinoacetyl)<br />

quinolin-2(1H)-one 60<br />

Girish Bolakatti, Manjunatha S. Katagi, S.N. Mamledesai, Sujatha M.L., Prakash Dabadi, Narayana Miskin.<br />

10.5530/rjps.2012.1.8<br />

• Evaluation of gastroprotective ability of Amorphophallus paeoniifolius corms against indomethacin<br />

induced gastric ulcers 67<br />

H.N. Nataraj, R.L.N. Murthy, Ramachandra Setty 10.5530/rjps.2012.1.9<br />

• Immunomodulatory activity of methanolic extracts of Pongamia glabra Vent. seeds and bark<br />

in cyclophosphamide induced mice 74<br />

Sanjeev Heroor, Arunkumar Beknal, Nitin Mahurkar 10.5530/rjps.2012.1.10<br />

• Simultaneous fi rst derivative UV spectrophotometric estimation of ramipril and olmesartan 78<br />

Santosh R. Karajgi, C.C. Simpi, Kalyane N.V. 10.5530/rjps.2012.1.11<br />

• Purifi cation and characterization of thermostable amylase from a strain of<br />

thermoactinomyces thalpophilus KSV 17 83<br />

K. Sreenivasa Rao, P. Ellaiah, Karnakumar V. Biradar 10.5530/rjps.2012.1.12


Knowing the known to understand the unknown<br />

A systematic approach to reviewing the scientifi c literature<br />

Yamuna V Kuberappa 1 , Arun HS Kumar 2<br />

1 Freelance MEMS Engineer, Cork, Ireland<br />

2 Department of Medicine (Cardiovascular), University College Cork, College Road, Cork, Ireland<br />

ABSTRACT<br />

Every work must have a starting point, and scientifi c research is no exception. Reviewing literature is an initial<br />

step in undertaking scientifi c research. Reviewing literature, as an ongoing process, provides a critical overview<br />

on the topic of interest and keeps the researcher up-to-date to pursue novel research plans. While reviewing<br />

literature helps early career investigators identify their research niche, established researchers review the literature<br />

to propagate the state of the art progress/opinions in their fi eld of expertise. Most of these reviews often guide<br />

the general research community. Various formats are often adopted in writing a review depending on the context,<br />

audience, depth/complexity of subject and extent of commercial value. The primary goal is to provide a critical<br />

and simplifi ed analysis of the facts. Essentially, every researcher should adapt to these formats in a unique way<br />

and should try to develop their own style of reviewing the literature. In this review, we will describe some of the<br />

essential steps to approaching various formats of reviewing the literature.<br />

Key words: Reviewing literature, scientifi c research, business reviews, book reviews<br />

INTRODUCTION<br />

Reviewing literature is not only a skill but<br />

also an art, wherein scientifi c knowledge<br />

and opinions are disseminated in a critical,<br />

concise and simplifi ed language to scientifi c<br />

and non-scientifi c communities. 1–16 The<br />

purpose of literature review is to provide<br />

critical opinions on established scientifi c<br />

facts and in the process evolve the strengths,<br />

weakness, opportunities and limitations on<br />

the topic of research interests (Figure 1).<br />

Hence, the emphasis is always on providing<br />

critical opinions on a topic rather than<br />

compiling scientifi c facts. 4,10,11,17–19 When one<br />

attempts to critically compile an opinion,<br />

it helps to identify gaps in the existing<br />

literature, and develop research questions<br />

for further investigation. Thus, the seed of<br />

research is sown. A researcher nurtures this<br />

seed over the course of his/her career, to<br />

grow into a fruitful tree. Before starting a<br />

review, clearly defi ne the objectives of the<br />

study and identify the targeted audience.<br />

Based on these two factors, decide the portal<br />

of publication for which the review would<br />

best fi t. The next step is to understand<br />

the procedure for searching and collecting<br />

materials for the intended work. This will<br />

be described in subsequent issues of this<br />

journal. This article focuses on general<br />

steps for systematically reviewing scientifi c<br />

literature.<br />

What necessitates the review<br />

of literature?<br />

Seeking information to gain understanding<br />

of any concept is a fundamental feature of<br />

learning. This is especially true in scientifi c<br />

research, where one needs to understand<br />

an established concept on a relevant topic<br />

to progress further, to evolve innovative<br />

ideas and thoughts, and to avoid any<br />

duplication. 2,6,10,18,22 The famous quote of ‘life<br />

begets life’ in the scientifi c research arena can<br />

Scien fi c Tools<br />

Received Date : 11-02-2012<br />

Revised Date : 29-02-2012<br />

Accepted Date : 03-03-2012<br />

DOI: 10.5530/rjps.2012.1.1<br />

Address for<br />

correspondence<br />

Arun HS Kumar, DVM, PhD.<br />

Senior Scientist and Head<br />

(Functional Biology)<br />

Department of Medicine<br />

(Cardiovascular)<br />

University College Cork<br />

Cork, Ireland<br />

E-mail: a.kumar@ucc.ie<br />

www.rjps.in<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 1


Figure 1: Six major reasons for reviewing the literature.<br />

be represented as ‘research begets research’ and ‘innovations’<br />

i.e., every research work contributes to further research<br />

bringing in scientifi c advancement and innovations.<br />

Therefore, the fundamental reason to proceed with<br />

reviewing the literature is to gain clear understanding on<br />

the topic of interest, so as to set the vision for future<br />

research and to provide the latest source of information<br />

on the topic to a wider scientifi c and non-scientifi c<br />

audience 10,16,25 (Figure 1). The overall objectives of<br />

writing a literature review are summarized in Figure<br />

2. There are several benefi ts of conducting scientifi c<br />

literature review (Box 1).<br />

The following questions are helpful before initiating a<br />

literature review.<br />

Why? Ask yourself, why do you want to review the<br />

literature on a specifi c topic? Come up with rational<br />

thinking on why a review of the literature is required on<br />

the topic at this time.<br />

Why not? This aspect is specifi cally relevant to<br />

young researchers, which emphasizes on curiosity and<br />

inquisitiveness. Hence, start reviewing the literature as<br />

a self-learning step to gain more insight on a specifi c<br />

scientifi c topic and ask yourself if it provokes interests<br />

or motivates you to proceed further.<br />

Box 1: Benefi ts in conducting a methodical literature<br />

review<br />

1. Gives a general and critical overview on the research topic<br />

2. Helps to improve the experimental methods and avoid<br />

duplication of research work<br />

3. Assists in identifying gaps in current research work<br />

4. Helps to build and defi ne research questions and<br />

hypotheses<br />

Yamuna V Kuberappa et al.: Knowing the known to understand the unknown<br />

Why not me? Question yourself on how well you are<br />

suited to reviewing the literature. This is more about<br />

setting the objectives based on strengths and weaknesses<br />

and defi ning targeted audience or scientifi c area of<br />

research.<br />

Why not me now? This question is more relevant to<br />

established researchers, wherein they initiate literature<br />

review to suite the requirement of the scientifi c<br />

community.<br />

The four why’s described above in the context of<br />

literature review might be applicable to any type of<br />

literature review and thus, readers are encouraged to<br />

follow them.<br />

How to start writing a review of literature?<br />

As stated previously, master the skills to search the<br />

literature. A major challenge is how to be effective in<br />

critically judging the published literature? With a recent<br />

surge in the number of scientifi c journals, the number<br />

of scientifi c manuscript/books published on any topic/s<br />

is on an exponential rise. Thus, vast and diversifi ed<br />

resources are available for researchers. It is often diffi cult<br />

to maintain the quality and standards uniform among all<br />

these resources; hence, the researchers should develop<br />

skills to critically evaluate the available literature. 6,9,10,26–28<br />

One of the ways to critique is by avoiding a researcher’s<br />

own inference based on the reported results and rather<br />

looking into the techniques by which the reported<br />

results were derived. Hence, it is emphasised that<br />

while reviewing any literature, one must be critical in<br />

interpretation whether the reported results are based on<br />

rigorous experimental designs or, are merely based on<br />

protocols, which are subjective, controversial and not<br />

rigorous. 10,23,27,28<br />

It is said ‘well begun is half done’; hence, it is important to<br />

start writing with a clear concept and focus by clearly<br />

identifying the objectives of the review. The best way<br />

Figure 2: The basic objectives of any literature review process.<br />

2 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


to do this is, by writing an abstract and clearly outlining<br />

headings and subheadings. 1,4,9,10,16,22,28,29 Once an outline<br />

is framed, start compiling the review by critically<br />

providing an opinion on the topic rather than merely<br />

summarizing the established facts. Attempt to identify<br />

various gaps in existing literature, such as an overworked<br />

area. Based on the existence of such gaps, write research<br />

questions. 10,28,29<br />

It is important to be creative in thoughts and to bring<br />

the researcher’s own signature style of formulating the<br />

literature review. Researchers should not hesitate to<br />

rationally challenge assumptions of existing scientifi c<br />

theory. The review should not be just a free fl ow of<br />

words with stagnant thoughts; rather it should be a<br />

free fl ow of critical thoughts and concepts which are<br />

innovatively constructed/reconstructed. 2,10,27,28,30 An<br />

important factor to innovation is being divergent in<br />

thoughts and attempting to think critically. A detailed<br />

review on critical thinking has recently been published<br />

in this journal, and readers are encouraged to read the<br />

article. 27 In order to draw critical insights from existing<br />

literature, it is necessary to follow an approach of slicing<br />

and dicing of the existing literature to highlight differing<br />

arguments, theories and methodologies. It often<br />

helps to summarize the existing research in the form<br />

of tables, key points on the box and/or fi gures rather<br />

than being extensively textual in literature review. Finally,<br />

it often helps to pre-identify the journal to which you<br />

intend to submit your literature review. This helps you<br />

to structure your review to fi t the aims and scope of<br />

the journal and plan your writing to meet the needs<br />

of specifi c audience.<br />

Defi ning the scope and formulating<br />

the literature review<br />

Composing of literature review begins with clearly<br />

defi ning the objectives based on nature of its intended<br />

audience and the degree of novelty (Figure 3). A wellcomposed<br />

literature review should comprehensively<br />

highlight the current state of the art on the topic,<br />

identify the areas of controversial and/or confl icting<br />

opinions and should hypothesize the topics that need<br />

future research focus. 10,29,30 Since several publications<br />

with either similar and/or contradictory opinions are<br />

being referenced, it is appropriate to use linker words<br />

and phrases such as similarly, in addition to, moreover, likewise,<br />

again (for studies with similar opinions) and albeit, on the<br />

contrary, however, on the other hand, conversely, nevertheless (for<br />

articles with contradictory opinions) while compiling<br />

a literature review. 10,31 It is also essential to include a<br />

summary paragraph at the end to highlight briefl y the<br />

major focus and opinions expressed in a literature review<br />

that often might link to a research hypothesis and future<br />

research directions. 10,22,31<br />

Yamuna V Kuberappa et al.: Knowing the known to understand the unknown<br />

Figure 3: Factors contributing to structuring literature review.<br />

The outline of any literature review is based on nature of audience<br />

and degree of novelty of the contents, which accordingly reflects<br />

its primary goal (creating awareness, educational or scientific<br />

advancement).<br />

Having familiarized with the general formula for<br />

composing a literature review, we now look into the<br />

practical recipes of literature review as it is very much<br />

context dependent. Every scientifi c literature review<br />

begins with the acquaintance of an existing literature and<br />

it is vital to identify type of materials to read. 2,10,18,23,28–30,32<br />

It often helps to categorize the existing literature based<br />

on the degree of novelty and similar versus contradictory<br />

opinions. Although these aspects are more relevant to<br />

established investigators, the novices are often directed<br />

by their guides/supervisors toward a list of introductory<br />

reference materials. While reading these introductory<br />

references, it is important to make notes and write<br />

critical comments as bullet points, which would come<br />

handy at the time of composing the literature review.<br />

The basic elements of a literature review include<br />

an introduction/background section highlighting<br />

the central theme and related context of the review.<br />

This is followed by the body of the literature review<br />

consisting of bulk of the text discussing the origin of<br />

the sources, theories and methods. Finally, the icing on<br />

the cake is the conclusions/recommendations section,<br />

a short paragraph consisting of critical opinion of the<br />

researcher on the subject matter and the scope for further<br />

research. Recently, a ‘Grounded Theory’ based approach<br />

(systematically adopting 5 stage processes) has been<br />

proposed for composing rigorous literature reviews 26,29<br />

(Figure 4), which is a signifi cant advancement of the<br />

matrix-based approach. 33 Although these approaches are<br />

primarily emphasized for information systems, general<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 3


Figure 4: The Grounded theory of composing literature review<br />

involves 5 sequential steps starting from defining to composing the<br />

literature review.<br />

concepts can be adopted for biological/life/medical<br />

sciences as well.<br />

Adopting a language style in composing<br />

literature review<br />

The major purpose of any language style is effective<br />

communication; hence, style is secondary, as long as<br />

writing is formal, clear and concise. 2,9,10,22,28,31 We highly<br />

encourage researchers to develop their own signature<br />

style, which is distinct from others. It is important to<br />

be objective rather than subjective to others opinions<br />

and always use present tense while referring to general<br />

opinions/theories and past tense while addressing specifi c<br />

study results. 10,31 Do not plagiarize any statements. One<br />

of the ways to avoid plagiarism is to make bullet points<br />

and reconstruct sentences to convey your opinion(s) in<br />

simple language. Remember, the essence of science is<br />

to make things as simple as possible. 10,31 Once a draft<br />

is done, it should be read several times and request a<br />

colleague to proofread. Incorporate all corrections and<br />

suggestions in the revised document.<br />

A literature review can take various formats depending<br />

on the purpose for which it is compiled. The following<br />

sections describe each of these formats.<br />

Composing the review of literature<br />

chapter for a thesis<br />

The major purpose of reviewing the literature for a<br />

thesis is to build the context on a research hypothesis<br />

on which the project is based. Thus, the subject matter<br />

should be organized around a pre-defi ned objectives and<br />

research parameters. 10,28,29,34 It often helps to organize<br />

Yamuna V Kuberappa et al.: Knowing the known to understand the unknown<br />

the structure of the literature review by compiling a<br />

list of clearly defi ned research questions/objectives<br />

and selectively reading the relevant literature to fi nd<br />

answers to these questions and in the process identify<br />

the gap in the knowledge base. 10,28,29,34 It is often easy<br />

to get side tracked; hence, keep focused on objectives<br />

and work toward formulating a clear thesis (research<br />

topic) statement and later propose a scientifi cally valid<br />

hypothesis. 7,10,16,20,22,25,26,28,29,34 Box 2 provides a checklist<br />

for compiling a literature review for a thesis.<br />

Organize literature review into a general outline<br />

consisting of introduction, body (major and minor<br />

topics) and conclusion sections. Follow the approach<br />

of composing rather then compiling, and wherever<br />

possible, summarize the data in the form of tables and/or<br />

fi gures. 7,10,16,20,22,25,26,28,29,34 Emphasise on why the intended<br />

work is important for the advancement of scientifi c fi eld<br />

and the likely benefi ts it intended to contribute. 7,10,28,29,34<br />

In doing so, comprehensively cover the existing<br />

literature within and closely related to the scientifi c area<br />

of investigation. Never avoid or ignore any scientifi c<br />

reports that are contradictory or controversial, rather<br />

acknowledge them with a comment why the reported<br />

views are different from those of the researcher. 10,28,34<br />

As noted previously, the review should be proofread<br />

by colleagues and friends who are from outside the<br />

scientifi c domain of the researcher as this clears blind<br />

spots and improves readability. 10,28,34 It is important<br />

that the literature review should lead the reader toward<br />

a conclusion that a rational and realistic research<br />

hypothesis is being proposed.<br />

For most thesis work, the sources are organized in a<br />

chronological pattern. For example, 10 if a researcher<br />

has decided to work on a novel stent-based therapeutics<br />

to achieve myocardial regeneration (MR) following<br />

myocardial infarction, he/she should start with background<br />

information on pathology of myocardial infarction and<br />

epidemiological data on the incidence of myocardial infarction<br />

world-wide and its socio-economic implications. The body of the<br />

review consists of, earlier therapeutic approaches, limitations of<br />

the existing therapeutic approaches and the concept of myocardial<br />

regeneration as a therapeutic option to treat myocardial infarction.<br />

BOX 2: Check list for the compilation of literature<br />

review for a thesis<br />

1. Does the literature review substantiate the research<br />

hypothesis?<br />

2. What is the key message delivered?<br />

3. Do you adequately cover the methodologies and techniques<br />

you intend to adopt?<br />

4. Have you adequately identifi ed the gaps and limitations in<br />

the knowledge base?<br />

5. Have you covered the existing literature up-to-date?<br />

4 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Follow this with a theory on why stents would be appropriate to<br />

deliver the therapeutics for myocardial regeneration. This should<br />

be followed by a discussion on the developments in design of stents<br />

and the era of introduction of drug eluting stents with its pros<br />

and cons. Furthermore, describe the limitations of current stent<br />

designs, the scope for advancement in the design of stents to deliver<br />

myocardial regenerative therapeutics and its futures perspectives.<br />

Finally, conclude with a statement why the reviewer approach<br />

is better than the other options available. (This paragraph is<br />

reproduced from our previous publication). 10<br />

Instructions for reviewing the literature for<br />

research or review articles<br />

Every research article consists of an introduction with<br />

a brief review of literature, which builds the context<br />

of the research work/hypothesis investigated. The<br />

purpose of literature review in a research article is to<br />

gain support for the scientifi c argument of a researcher.<br />

The purpose in the context of a review article, is to<br />

generate views by composing and synthesizing the<br />

arguments and ideas of others. 1,9,10,18,28,29,35,36 Both<br />

research and review articles have the same elements<br />

of literature but differ in terms of elaboration and<br />

discussion. In research articles, the literature review is<br />

integrated into the introduction and discussion section<br />

and the emphasis is on supporting a researcher’s<br />

argument. On the other hand, in a review article,<br />

the literature review is emphasised on background<br />

information and identifying gaps in knowledge base to<br />

build a critical opinion on the subject topic and future<br />

research directions. 2,9,10,29,36<br />

For example, if one discovers a novel stent design for<br />

treating coronary artery disease (CAD) and report the<br />

same in a research article, the following pattern may<br />

be used.<br />

‘Stents are a widely used medical device for treating CAD…. The<br />

bare metal and drug-eluting stents, which are extensively used,<br />

have serious drawbacks with respect to developing late stenosis<br />

and thrombosis complications … Several studies … report the<br />

complications associated with the currently available stents. Critical<br />

evaluation of the pathology …has revealed that complications are<br />

a consequence of the unique design of the stents. To overcome this<br />

limitation we have designed a novel stent, which we foresee to be<br />

devoid of the side effects observed with the currently available stent<br />

designs,…and tested it for performance in the current study with<br />

the following objectives’. (This paragraph is reproduced from<br />

our previous publication). 10<br />

While all the general guidelines for composing a<br />

literature review are applicable to research as well as<br />

review articles, it is acceptable and essential to include<br />

the reviewer’s opinions in the review articles, especially<br />

on the methodologies adopted and the robustness of<br />

the conclusions reported. Several categories of review<br />

articles exist (comprehensive review, meta-analysis, brief<br />

Yamuna V Kuberappa et al.: Knowing the known to understand the unknown<br />

review, hypothesis-based review, opinion article, stateof-the-art-review,<br />

etc.) depending on the scientifi c insight<br />

and extent of focus on the subject by the reviewer. For<br />

instance, the above paragraph in the context of a review<br />

article can be redesigned as follows.<br />

Start with a brief introduction on CAD, its incidence, its impact<br />

on human health and its socio-economic consequences. Follow this<br />

with a discussion on why are stents the obvious choice for CAD,<br />

what was the approach adopted before the introduction of stents<br />

with its pros and cons, how has the use of stents infl uenced the<br />

CAD events. Include a few paragraphs on the nature/design of<br />

stents used initially and the developments over time on the stent<br />

design and coatings. Follow this with one or two paragraphs on<br />

the types of stents currently in use, their limitations and commonly<br />

encountered side effects (critically evaluate these studies as they<br />

form the basis for further investigations in stent designing and<br />

coatings). Include a paragraph on the reviewer’s opinions for the<br />

limitations encountered with the use of currently available stents<br />

for CAD and the future strategies to overcome these limitations.<br />

Finally, conclude with the room available for future advancement/<br />

development. (This paragraph is reproduced from our<br />

another publication). 10<br />

Art of literature review for books/book chapters<br />

With the growth in digital media and the rising<br />

popularity of tablet devices/computers, the demand<br />

for e<strong>Book</strong>s has surged. <strong>Book</strong>s/book chapters remain a<br />

vital source of established knowledge to both scientifi c<br />

and non-scientifi c audiences. 2,10,18,29 Unlike research<br />

articles, the language used in books must be simple<br />

(avoiding scientifi c jargons wherever possible) and<br />

comprehensible to a wide audience. 10,29 In reviewing a<br />

topic for a book, one should limit to including established<br />

facts and present it with adequate diagrams, tables and<br />

fl ow charts to simplify concepts. It is often essential to<br />

adopt a narrative approach rather than validate scientifi c<br />

facts. Additionally, summarize each subsections, as this<br />

greatly improves readability and comprehension of<br />

the topic. 10,29<br />

Literature review for grant applications and<br />

research proposals<br />

The general outline and focus of the literature review<br />

for grant applications and research proposals are similar<br />

to the one outlined above. However, the emphasis is<br />

more on novelty and applicability aspects of research<br />

proposed and it is important to be precise and to the<br />

point. 10,22,28,29,37 More importantly, the applicant’s own<br />

research to the progress of the proposed project<br />

should be included in the application. This would instill<br />

confi dence in grant reviewers on the abilities and skills<br />

of the researcher to perform the work as described in<br />

the research proposal. 10,28,37 Such confi dence build-up<br />

measure among reviewers helps achieving high rating and<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 5


hence, the success of the grant application. As subject<br />

experts often review the grants/research proposals, it<br />

is necessary to highlight scientifi c advancement rather<br />

than basic information and scientifi c novelty. Needless<br />

to mention that the literature review supporting the<br />

scientifi c advancement must always be tailored to meet<br />

the scope and objectives of the funding agency.<br />

Writing literature review for business proposals<br />

Increasing trends in the academic-industrial partnerships<br />

and academic spin-offs have lead to many academicians<br />

adopting business and entrepreneurship style in running<br />

their research labs. 10,28 Such an integration of corporate<br />

atmosphere in academic environment has progressively<br />

introduced the concept of academic business proposals,<br />

which often involve academic partnerships with<br />

industries to capitalize on the basic/applied science<br />

research to achieve mutual benefi ts. 10,28 Business<br />

proposals by academic researchers should focus on the<br />

following elements.<br />

1. Commercial benefi ts and the economics of the<br />

research work in question.<br />

2. Critical reviews of the applicant’s research work<br />

within the context of commercialization, novelty<br />

and patentability over related research competitors.<br />

Hence, unlike literature reviews for scientifi c manuscripts,<br />

grant proposals, and books, the write-up for business<br />

proposals should highlight on the economic and<br />

commercialization impact of the proposed work with<br />

direct links to novelty and patentability.<br />

SUMMARY<br />

Literature reviews are a valuable knowledge source and a<br />

measure of consolidating the current research progress<br />

in any scientifi c fi eld. There are several categories of<br />

review articles, e.g., comprehensive reviews, metaanalysis,<br />

brief reviews, opinion articles, etc. All literature<br />

reviews adopt the same broad outline/structure, but<br />

vary in detail depending on the objectives. In general,<br />

these objectives identify gaps in existing work, avoid<br />

duplication of work, and propose a scientifi cally valid<br />

hypothesis. Depending on the context in which it is<br />

composed, a literature review has many benefi ts. It may<br />

be a source of self-education or educating others and<br />

publication. In addition, reviews attract successful grant<br />

funding and might build a commercialization case for a<br />

body of research. We emphasise that literature reviewing<br />

is an art of scientifi c communication, which can, if<br />

properly routed, lead to intellectual questions and grants<br />

or business opportunities. Hence, it is necessary to invest<br />

considerable amounts of time, effort and dedication to<br />

this vital scientifi c activity.<br />

Yamuna V Kuberappa et al.: Knowing the known to understand the unknown<br />

CONFLICT OF INTEREST: None.<br />

REFERENCES<br />

1. Bruce J, Mollison J. Reviewing the literature: adopting a systematic<br />

approach. J Fam Plan Reprod Health Care 2004;30:13–16.<br />

2. Connelly LM. Reviewing the literature. Medsurg Nurs. 2010;19:<br />

245–246.<br />

3. Davis EA, Bagley TC, Panacek EA. Basics of research (Part 2):<br />

Reviewing the literature. Air Med J 1995;14:101–105.<br />

4. Epstein E. Reviewing the medical literature: statistics alone are not<br />

enough. Arch Dermatol 2002;138:829–830.<br />

5. Gray M. Reviewing the literature. J Wound Ostomy Continence Nurs<br />

1995;22:71–72.<br />

6. Griffi ths P, Norman I. Science and art in reviewing literature. Int J Nurs<br />

Stud 2005;42:373–376.<br />

7. Hendry C, Farley A. Reviewing the literature: a guide for students. Nurs<br />

Stand 1998;12:46–48.<br />

8. Horvath AR, Pewsner D. Systematic reviews in laboratory medicine:<br />

principles, processes and practical considerations. Clinica Chimica<br />

Acta 2004;342:23–39.<br />

9. Isaacson RJ. Reviewing the literature traditional, systematic, evidencebased.<br />

Angle Orthod 2005;75:888–889.<br />

10. Kumar AHS. Comprehensive review of the literature. In: Jagadeesh<br />

G, Murthy S, Gupta YK, Prakash A, editors. Biomedical research: from<br />

ideation to publication. New Delhi: Wolters Kluwer Health, Lippincott<br />

Williams & Wilkins 2010:377–387.<br />

11. Luczun ME. Reviewing the literature: a treasure hunt. J Post Anesth<br />

Nurs 1987;2:120–122.<br />

12. McNally R, Alborz A. Developing methods for systematic reviewing in<br />

health services delivery and organization: an example from a review<br />

of access to health care for people with learning disabilities. Part 1.<br />

Identifying the literature. Health Info Lib J 2004;21:182–192.<br />

13. Sharts-Hopko NC. Reviewing the literature. J Assoc Nurses AIDS Care<br />

1998;9:87–89.<br />

14. Sheehan J. Research. 2. Reviewing the literature. Nurs Mirror 1985;<br />

160:29–30.<br />

15. Summers S. Steps in reviewing literature. J Post Anesth Nurs 1991;<br />

6:188–192.<br />

16. Webster J, Watson RT. Analyzing the past to prepare for the future: writing<br />

a literature review. MIS Quarterly 2002;26:13–23.<br />

17. Williams HA. Searching the literature creatively: updating your skills in<br />

reviewing the literature. J Pediatr Oncol Nurs 1993;10:33–36.<br />

18. Coppola N, Carliner S. Is our peer-reviewed literature sustainable?<br />

Prof Communication Conf IEEE Int 2011;17–19 Oct:1–7. DOI: 10.1109<br />

/IPCC.2011.6087222.<br />

19. MacGuire J. Reviewing the research literature. Nurs Stand 1990;4:35–36.<br />

20. Bell FJ. Reviewing the literature: a student’s perspective. J Child Health<br />

Care 1998;2:122–127.<br />

21. Koziol-McLain J, Tanabe P. Reviewing the research literature: you don’t<br />

have to do it alone. J Emerg Nurs 1996;22:352–355.<br />

22. Price B. Guidance on conducting a literature search and reviewing<br />

mixed literature. Nurs Stand 2009;23:43–49.<br />

23. Sherwin T, Foggin SM, Cartwright VA. Finding information in medical<br />

sciences: Searching the medical Literature. In: Jagadeesh G, Murthy<br />

S, Gupta YK, Prakash A, editors. Biomedical research: from ideation<br />

to publication. New Delhi: Wolters Kluwer Health, Lippincott Williams &<br />

Wilkins 2010:388–408.<br />

24. Schwartz RB, Russo MC. How to quickly fi nd articles in the top IS journals.<br />

Commun of the ACM 2004;47:98–101.<br />

25. Swanson JM, Easterling P, Costa L, et al. Student-staff collaboration in<br />

identifying nursing problems and reviewing the literature. West J Nurs<br />

Res 1990;12:262–266.<br />

26. McGhee G, Marland GR, Atkinson J. Grounded theory research: literature<br />

reviewing and refl exivity. J Adv Nurs 2007;60:334–342.<br />

27. Reisman F. Creative, critical thinking and logic in research. RGUHS<br />

J Pharm Sci 2011;1:97–102.<br />

28. Sandberg J, Alvesson M. Ways of constructing research questions:<br />

gap-spotting or problematization? Organization 2011;18:23–44.<br />

29. Wolfswinkel JF, Furtmueller E, Wilderom CPM. Using grounded theory<br />

as a method for rigorously reviewing literature. Eur J Inform Syst<br />

2011: advance online publication 29 November 2011; doi: 10.1057<br />

/ejis.2011.51;1–11.<br />

6 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


30. Franks H, Hardiker NR, McGrath M, et al. Public health interventions<br />

and behaviour change: Reviewing the grey literature. Public Health<br />

2012;126:12–17.<br />

31. Prakash A. ABC of medical writing: Language and style in scientifi c<br />

communication. In: Jagadeesh G, Murthy S, Gupta YK, Prakash A,<br />

editors. Biomedical research: from ideation to publication. New Delhi:<br />

Wolters Kluwer Health, Lippincott Williams & Wilkins 2010:421–427.<br />

32. Srikumar BN. Reference style and common errors with referencing.<br />

In: Jagadeesh G, Murthy S, Gupta YK, Prakash A, editors. Biomedical<br />

research: from ideation to publication. New Delhi: Wolters Kluwer<br />

Health, Lippincott Williams & Wilkins 2010:479–488.<br />

33. Salipante P, Notz W, Bigelow J. A matrix approach to literature reviews.<br />

Res Organ Behav 1982;4:321–348.<br />

Yamuna V Kuberappa et al.: Knowing the known to understand the unknown<br />

34. Hardy S, Ramjeet J. Refl ections on how to write and organise a research<br />

thesis. Nurse Res 2005;13:27–39.<br />

35. Thomas BH, Ciliska D, Dobbins M, et al. A process for systematically<br />

reviewing the literature: providing the research evidence for public<br />

health nursing interventions. Worldviews Evid Based Nurs 2004;1:<br />

176–184.<br />

36. Humphery SE. What does a great meta-analysis look like? Organ Psychol<br />

Rev 2011;1:99–103.<br />

37. Gulati A. Writing Grant Proposal. In: Jagadeesh G, Murthy S, Gupta<br />

YK, Prakash A, editors. Biomedical research: from ideation to publication.<br />

New Delhi: Wolters Kluwer Health, Lippincott Williams & Wilkins<br />

2010:519–526.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 7


Pharmaceutical discovery and development<br />

(A report on the symposium held in 63rd annual conference<br />

of the Indian Pharmaceutical Congress-2011)<br />

Pitchai Balakumar 1 , Gowraganahalli Jagadeesh 2,§<br />

1 Department of Pharmacology, Institute of Pharmacy, Rajendra Institute of Technology and Sciences, Sirsa 125055,<br />

Haryana, India<br />

2 Division of Cardiovascular and Renal Products, Center for Drug Evaluation and Research, US Food and Drug Administration,<br />

Silver Spring, MD 20993, USA<br />

ABSTRACT<br />

The 63rd Annual conference of the Indian Pharmaceutical Congress (IPC) held at Bangalore International Exhibition<br />

Centre, Bengaluru, Karnataka, India, from 16 to 18 December, 2011, was organized by ‘Indian Pharmaceutical<br />

Congress Association’ and hosted by ‘The Indian Pharmaceutical Association’, Karnataka State Branch. The<br />

conference included a 3-hour symposium on “Pharmaceutical Discovery and Development”. The objectives of this<br />

symposium were to enlighten the delegates on a few key aspects of drug discovery and development. To that<br />

end, the symposium addressed on drugs and drug targets, early evaluation of pharmacokinetics and metabolism<br />

in drug discovery, toxicity tests in drug development, regulatory review process, and biomarkers.<br />

INTRODUCTION<br />

Drugs are miracles of modern science. They<br />

are used to prevent, treat, or cure diseases<br />

and medical conditions. Disease processes<br />

are often complex and involve a defi ned<br />

sequence of events. Drugs are designed to<br />

intervene in the disease process. They can<br />

simply alleviate symptoms or even control<br />

subsequent development of a chronic<br />

disease condition. Except for antimicrobials<br />

and to some extent anti-infl ammatory and<br />

anti-neoplastic drugs, most other drugs<br />

infrequently cure diseases. Yet, these<br />

drugs improve the quality of life and, in<br />

general, a sense of well-being. Billions of<br />

prescriptions are fi lled worldwide every year.<br />

The process of discovering, developing<br />

and testing new drugs encompasses some<br />

of the most inspiring areas of scientifi c<br />

discovery. 1 Drug development involves<br />

multiple disciplines encompassing a large<br />

number of skilled scientists at different<br />

levels, interdependent science managers<br />

involved in important decision-making,<br />

huge volumes of generated data, and<br />

ultimately the drug regulatory agencies (e.g.,<br />

US FDA, European Medicines Agency,<br />

Japanese Ministry of Health and Welfare). 1<br />

It is a complicated, time-consuming (often<br />

>10 years), and costly process (over US $1<br />

billion) whose end result is seldom known<br />

at the outset. 2<br />

The initial stages of drug discovery<br />

research are characterized by target discovery<br />

(identifi cation and validation) and lead<br />

discovery (generation and optimization).<br />

Integration of combinatorial chemistry,<br />

high throughput screening, and molecular<br />

§ The opinions expressed herein are those of GJ and do not necessarily refl ect those of the US Food and Drug<br />

Administration.<br />

Mee ng Report<br />

Received Date : 15-02-2012<br />

Revised Date : 27-02-2012<br />

Accepted Date : 02-03-2012<br />

DOI: 10.5530/rjps.2012.1.2<br />

Address for<br />

correspondence<br />

Gowraganahalli<br />

Jagadeesh, PhD.<br />

Senior Expert Pharmacologist<br />

Division of Cardiovascular and<br />

Renal Products<br />

Food and Drug<br />

Administration (FDA)<br />

10903 New Hampshire Ave<br />

Bldg 22, Rm 4128<br />

Silver Spring, MD<br />

20993-0002, USA.<br />

Phone: 001-301 796 1093<br />

Fax: 301-796-9838<br />

E-mail: gowra.jagadeesh@fda.<br />

hhs.gov (G Jagadeesh)<br />

pbala2006@gmail.com<br />

(P Balakumar)<br />

www.rjps.in<br />

8 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


modeling generates an abundance of candidate molecules<br />

with drug-like properties. The lead compounds<br />

are selected based on a high degree of selectivity for<br />

the target. Such compounds are also optimized in terms<br />

of potency, physicochemical properties, pharmacokinetics<br />

and drug metabolism, and toxic profi le. 3,4 This<br />

information helps to reduce attrition of compounds<br />

and contributes markedly to success rates in identifying<br />

lead candidates. With only additional minor changes in<br />

structure or other physico-chemical properties of the<br />

lead compound, a potentially viable drug candidate is<br />

launched (Box 1).<br />

The quality of the lead compound is crucial to the<br />

success of the refi nement and development process. The<br />

lead drug candidate that enters the initial developmental<br />

stage (see Box 1) must have a strong chance of meeting<br />

the target profi le in terms of effi cacy, safety, route of<br />

administration, and treatment regimen. An early step of<br />

drug development includes pharmacologic, toxicologic,<br />

pharmacokinetic, and metabolism/biotransformation<br />

studies both in vitro and in animals. These studies<br />

should reveal potentially useful (intended target) and<br />

harmful (off target) properties of the drug candidate<br />

in a qualitative and quantitative manner, thus permitting<br />

an assessment of therapeutic risk. An investigational<br />

new drug (IND in the USA) application is then fi led<br />

with the drug regulatory agency demonstrating that<br />

the CMC (chemistry and manufacturing controls),<br />

nonclinical pharmacologic and toxicologic properties<br />

of the drug have been well characterized. The next<br />

step in drug development (clinical phase) is to<br />

demonstrate that the benefi t-to-risk ratio as observed<br />

in pharmacology and toxicology studies is confi rmed<br />

Box 1: The Discovery to Development Scheme<br />

1. Drug discovery (Early pharmaceutical research)<br />

A. Target discovery (Exploratory phase)<br />

1. Target identifi cation<br />

2. Target validation<br />

B. Hits discovery<br />

1. Hits identifi cation<br />

2. Hits validation (after hits reproducibility and dose effect)<br />

C. Lead discovery<br />

1. Lead generation/identifi cation<br />

2. Lead refi nement/optimization<br />

2. Drug development (Late Pharmaceutical R & D; drug<br />

candidate launched)<br />

A. Drug candidate selection<br />

B. Preclinical development (short and long term studies)<br />

C. IND application fi led<br />

D. Clinical development (Phase 1, 2, 3)<br />

E. NDA fi led<br />

F. Drug regulatory agency approval for marketing<br />

Balakumar, Jagadeesh: Pharmaceutical Discovery and Development<br />

in patients. Standard endpoints in animal safety studies<br />

focus upon morphologic assessments of tissues,<br />

whereas the end points in initial human safety studies<br />

(Phase 1) are largely physiologic, including assessments<br />

of the cardiovascular system, central nervous system,<br />

respiratory, and renal functions. At the completion of<br />

phase 2 and phase 3 trials, if conclusive evidence of the<br />

new molecule’s safety and effectiveness for a specifi c<br />

indication exists, a new drug application (NDA in the<br />

USA) is submitted to the drug regulatory agency. If<br />

benefi ts of a drug for the intended patient population<br />

outweigh the risks, the drug will receive approval from<br />

the regulatory agency and thus, can be marketed (Box 1).<br />

An overview of the symposium: This symposium addressed<br />

selected key topics in drug discovery and development,<br />

such as potential drug targets for therapeutic interventions,<br />

early evaluation of pharmacokinetics and drug<br />

metabolism, preclinical toxicity studies, and regulatory<br />

guidelines in submission of investigative and new<br />

drug applications. It concluded with the role of novel<br />

and traditional biomarkers in monitoring disease or its<br />

treatment during the course of clinical trials. The purpose<br />

of this report is to highlight and disseminate key<br />

information presented at the symposium as a means of<br />

educating the scientifi c community so that they can stay<br />

current with this rapidly developing fi eld.<br />

The symposium had fi ve distinguished speakers affi liated<br />

to drug regulatory affairs, industries and academia<br />

from India and the USA. The symposium was arranged<br />

by Dr. G. Jagadeesh of the US Food and Drug Administration<br />

(FDA), and chaired by Prof. P. Balakumar of the<br />

Institute of Pharmacy, Rajendra Institute of Technology<br />

and Sciences, Sirsa, Haryana, India.<br />

Drug Targets<br />

The symposium began with a welcome address by<br />

Dr. H.G. Shivakumar of J.S.S College of Pharmacy,<br />

Mysore, Karnataka. Prof. P. Balakumar, the chairman<br />

of the symposium, briefl y overviewed the topics for<br />

presentation and profi led the speakers. Dr. G. Jagadeesh<br />

(US FDA) then delivered the fi rst lecture on ‘Drugs and<br />

Drug Targets’. A main element of early pharmaceutical<br />

research is to reveal potential drug targets for therapeutic<br />

intervention in the intended patient population.<br />

Determination of the optimal molecular targets for<br />

drug intervention provides the basis for the discovery<br />

of new medicines. Disease-relevant target identifi cation<br />

is considered an important criterion for determining<br />

the correlation between drug and indication.<br />

An important focus of drug discovery has been on<br />

protein drug targets as a means of treating various<br />

disorders. The advent of genomic sciences has<br />

advanced our knowledge in the development of new<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 9


pharmaceuticals, as we now have a better understanding<br />

of how genes are linked to diseases. It is estimated that<br />

approximately 10% of the 25,000–30,000 known human<br />

genes are linked to diseases. Identifying individual genes<br />

considered potential targets (i.e., drugome) is not an ideal<br />

task, because the functional product of each gene or<br />

proteome is under multiple regulatory controls, including<br />

splice variants and post-translational modifi cations.<br />

Thus, targeting the body’s proteins or proteome is ideal<br />

because druggability, or the ability of a drug to affect<br />

a protein’s function, may be an inherent property of a<br />

given protein. However, this does not constitute a useful<br />

drug target unless the protein is also linked to a disease.<br />

Nearly half of the disease-associated proteins are not<br />

druggable as they lack a hydrophobic binding site for<br />

a potential orally-delivered, cell membrane-permeable<br />

drug molecule. This restricts the number of potential<br />

drug targets to less than 1500. 5 Further, protein drug<br />

targets should be viewed in the context of various<br />

interrelated networks involved in the disease process.<br />

Various factors such as genetic mutations, epigenetic<br />

changes and pathogens can cause perturbations in the<br />

function of networks, disrupting signaling pathways and<br />

resulting in diseases. According to the Online Mendelian<br />

Inheritance in Man, more than 1284 distinct disorders<br />

have been reported. These are classifi ed into 22 disorder<br />

classes based on the physiology of the system and 1777<br />

disease-related genes. 6 Although the majority of disease<br />

genes and the molecular basis for these diseases are not<br />

known, knowledge of the human genome sequence has<br />

increased potential new targets. The effective number of<br />

novel drug targets has been calculated by the intersection<br />

of the number of druggable proteins with the number<br />

of disease-associated proteins, or 600–1500 novel drug<br />

targets (2–5% of the genome). 5,7<br />

Noting the report of Rask-Andersen et al., 8<br />

Dr. Jagadeesh said that for the past thirty years (1982<br />

to 2010) of drug target innovation, on average, about<br />

18 new drugs targeting human proteins have been<br />

approved by the FDA every year, of which about 4<br />

are fi rst-in-class that act on novel target structures that<br />

are encoded by the human genome, and the remaining are<br />

follower or ‘me-too’ drugs. 8 According to these authors,<br />

this translates to 435 drug targets over that time period.<br />

Enzymes are the largest target class (47%) followed by<br />

receptors (G-protein coupled receptors account for 30%,<br />

the second largest), transporters (voltage-gated calcium<br />

channels, the largest) and others. The most common<br />

pharmacologic action of the approved drugs is antihypertensive<br />

followed by anti-cancer, anti-infl ammatory,<br />

sedative and hypnotic, anti-allergic, anti-convulsant, antiarrhythmic,<br />

anti-psychotic, anti-depressant and analgesic.<br />

Most drugs target only a select number of proteins<br />

but some can target as many as 19 (e.g., bromazepam).<br />

Balakumar, Jagadeesh: Pharmaceutical Discovery and Development<br />

The average number of target proteins reported per drug<br />

is 1.8 surpassing the number of single target drugs, 6 and<br />

thus, are called target multipliers (polypharmacology).<br />

Multi-target drugs are useful in cases where a<br />

specifi c disease-associated protein is not druggable.<br />

Dr. Jagadeesh also discussed various interactions, such as<br />

between proteins (interactome), drugs (drug network),<br />

drug and target (drug-target network) and disease and<br />

disease genes (diseasome). It is anticipated that analysis<br />

of these complex networks may also lead to new avenues<br />

for drug target prediction.<br />

Early Pharmacokinetics and Drug Metabolism<br />

After lead molecules are identifi ed, they are optimized<br />

in terms of potency, selectivity, ADME (absorption,<br />

distribution, metabolism, and excretion) and toxicology<br />

before they can become candidates for further drug<br />

development. 3,9 It is important to emphasize the<br />

integral role of drug metabolism and pharmacokinetics<br />

(DMPK) in early phases of drug discovery, so as<br />

to avoid failure due to poor pharmacokinetics and<br />

bioavailability that largely result from suboptimal<br />

physico-chemical properties. 10 DMPK scientists play<br />

a multifaceted role in various functional areas, such as<br />

drug biotransformation, pharmacology and toxicology<br />

that help provide valuable information related to effi cacy<br />

and safety of a new molecule. Dr. Sandhya Mandlekar<br />

(Bristol-Myers Squibb India Ltd., India) spoke on the<br />

topic of ‘Pharmacokinetics and Metabolism in Early<br />

Drug Discovery’. While defi ning the evolving role<br />

of DMPK studies in early pharmaceutical research,<br />

Dr. Mandlekar described traditional ADME properties,<br />

such as characterizing bioavailability and half-life in<br />

animals and man. She also discussed advances in the fi elds<br />

of pharmacogenetics, pharmacogenomics and drug<br />

transporters; the shift in drug metabolism paradigms,<br />

such as enzyme regulation, drug-drug interaction and<br />

inter-individual variability; and predictive biomarkers<br />

from animals to man. She also described the availability<br />

of new technologies and using a tiered approach to<br />

DMPK studies. Her presentation also focused on in vitro<br />

models to predict hepatic clearance, CYP inhibition and<br />

induction assays, Caco-2 permeability determination<br />

assay for poorly soluble compounds, generating<br />

in vivo EC50s by modeling nonclinical PK/PD data, and<br />

investigating unusual metabolic reactions. Dr. Mandlekar<br />

also addressed necessary steps in early pharmaceutical<br />

research ADME studies (Box 2).<br />

Finally, Dr. Mandlekar suggested that the mission of<br />

discovery ADME studies is to ensure no development<br />

candidate fails in the clinic due to unforeseen ADME<br />

properties, and to improve developability characteristics<br />

and risk assessment, in parallel with effi cacy, to get better<br />

success rates during clinical development.<br />

10 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Box 2: A Focused Application of Discovery ADME<br />

Studies<br />

1. Active involvement earlier in the Discovery Process<br />

2. Timely guidance to Chemistry to select chemotypes with<br />

desirable ADME properties<br />

3. Maximize informed decision making during Lead<br />

Optimization<br />

4. Improved ability to predict human DMPK<br />

5. Stronger partnership with all areas of Drug Discovery and<br />

Development<br />

Toxicology Studies<br />

A remaining critical issue in drug development is the<br />

failure to identify, suffi ciently, toxicological problems<br />

pertaining to new chemical entities early enough in the<br />

drug research and development process. Better success<br />

in developing early toxicologic screens will certainly<br />

enhance the number of successful new chemical entities<br />

entering to the clinical developmental phases and, in<br />

fact, avoid the expense of terminating drugs in latestage<br />

clinical development. Toxicity is a leading cause<br />

of attrition at both clinical and nonclinical stages of<br />

drug development. 4,10 The primary objective of nonclinical<br />

toxicology studies in the drug discovery process<br />

is to evaluate the safety of potential drug candidates for<br />

human use. The ultimate goal is to translate the in vitro<br />

and animal responses into an understanding of the risk<br />

for human subjects under therapeutic conditions. Thus,<br />

non-clinical toxicology testing is utilized at all stages of<br />

the drug discovery and development processes.<br />

Dr. K.S. Rao (Rao Toxicology Foundation, Bengaluru,<br />

India, and Editor-in-Chief of Toxicology International)<br />

spoke at length on ‘Toxicology Issues in Drug<br />

Development’. Dr. Rao addressed various toxicological<br />

issues encountered during the early phases of research<br />

and drug development. The objectives of toxicity<br />

tests, whether conducted in the whole animal or in<br />

cell systems, should be to predict the adverse effects<br />

of the tested compound in human beings both in a<br />

qualitative and quantitative manner. While in silico and<br />

in vitro models will continually be developed and refi ned,<br />

in vivo preclinical safety models remain as the gold<br />

standard for assessing human risk. Various cell culture<br />

systems are an in vitro model of a target tissue in the<br />

human body and mimic the response of human cells<br />

to drug exposure. Several differentiated cell types have<br />

been used in toxicology investigations. Among them,<br />

the more widely used are cells derived from liver, lung,<br />

heart, kidney, muscle, nervous and reticulo-endothelial<br />

systems. Primary cultures or isolated cells are widely<br />

used because the cells retain their specialized functions.<br />

Dr. Rao also discussed the rapid development of<br />

Balakumar, Jagadeesh: Pharmaceutical Discovery and Development<br />

‘Functional genomics’, which includes proteomics and<br />

transcriptomics, as an emerging discipline that represents<br />

a global and systematic approach to identifying biological<br />

pathways and processes in both normal and abnormal<br />

physiological states. Although numerous tests have been<br />

validated in the recent past, they not yet gained wide<br />

spread use in discovery toxicology.<br />

Dr. Rao noted that the fi rst of two stages in new drug<br />

development is comprised of non-clinical (pharmacology<br />

and toxicology) studies. The second stage is initiation<br />

of clinical studies and completion of long-term animal<br />

toxicity studies. The objectives of non-clinical studies<br />

are to demonstrate the biologic activity (effi cacy and<br />

selectivity) against the targeted disease and to provide<br />

data for toxicity and safety evaluation (risk assessment).<br />

Characterization of toxic effects with respect to target<br />

organs, dependence of dose to effect, relationship to<br />

systemic drug exposure, and potential reversibility of<br />

any toxicities are all helpful in human dose escalation<br />

strategies. The toxicity studies are limited at the beginning<br />

of clinical development, but additional long-term toxicity<br />

studies are conducted in tandem with on-going clinical<br />

studies to help support the intended dose and duration<br />

of the clinical trials. Serious adverse effects noted in<br />

ongoing clinical trials may call for additional toxicologic<br />

studies or even result in the clinical trials being halted<br />

(clinical hold).<br />

International regulatory guidelines require that new drugs<br />

be tested for safety and effi cacy before marketing. The<br />

recommendations for the extent of non-clinical safety<br />

studies to support various stages of clinical development<br />

differ only slightly among the regions of Europe, USA<br />

and Japan. International Conference on Harmonization<br />

(ICH) guidelines (ICH-M3) describe the consensus that<br />

exists regarding the scope and duration of non-clinical<br />

safety studies to support the conduct of human clinical<br />

trials for medicinal products. The major toxicology<br />

studies that are conducted to demonstrate the safety of a<br />

Box 3: Major Categories of Toxicology Studies<br />

Conducted During Drug Development (in Tandem<br />

with Clinical Phases)<br />

(http://www.fda.gov/Drugs/GuidanceCompliance<br />

RegulatoryInformation/Guidances/ucm065007.htm)<br />

1. Type, duration, and timing of non-clinical studies in support<br />

of clinical trials (ICH M3(R2))<br />

2. Safety Pharmacology studies (ICH S7A and S7B)<br />

3. Toxicokinetics (ICH S3A)<br />

4. Single and Repeat dose toxicity studies (ICH S4A)<br />

5. Genotoxicity studies (ICH S2(R1))<br />

6. Carcinogenicity studies (ICH S1A, S1B, S1C(R2))<br />

7. Reproductive and development toxicity (ICH S5A, S5B)<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 11


new drug candidate are outlined below (Box 3). Besides<br />

ICH, three other guidelines are available. Each of these<br />

describes methodology and assay criteria for most of the<br />

toxicology studies. They are: i) CFSAN Redbook 2000:<br />

Toxicological Principles for the Safety Assessment of<br />

Food ingredients; 11 ii) OECD guidelines; 12 and iii) EPA<br />

guidelines for toxicity testing. 13<br />

Regulatory Review Process in New Drug<br />

Development<br />

The long road from original concept to fi nal drug approval<br />

has multifaceted phases that include pharmacologic,<br />

pharmacokinetic and drug metabolism, toxicologic and<br />

clinical studies. Dr. Gopi Vudathala (Global Regulatory<br />

Affairs-Interface CMC Sanofi -Aventis, USA) spoke on<br />

‘Overview of the Drug Development Process: The<br />

IND and NDA Process’. His talk focused on the drug<br />

development process describing the regulations and<br />

guidances specifi c to the IND and NDA process. These<br />

included pre-IND, IND application and types of reviews,<br />

various phases of clinical trials, timing and duration of<br />

non-clinical studies necessary to support clinical trials,<br />

NDA registration process and necessary submission<br />

requirements, NDA review process, and fi nally, NDA<br />

actions. Dr. Vudathala integrated many of the issues<br />

with strategic preclinical and clinical drug development<br />

to the regulatory decision involved in licensing the<br />

product for marketing. He also spoke on ‘relevant Laws’<br />

(Federal Food, Drug, and Cosmetic Act; Public Health<br />

Service Act-Part F Licensing of Biological Products and<br />

Clinical Laboratories) and ‘relevant regulations’ (IND<br />

regulations (both drugs and biologics) 21 CFR 312.23;<br />

NDA (drugs) regulations-21 CFR 314.50; Product<br />

licensing (biologics)-21 CFR 601; Protection of human<br />

subjects and informed consent regulations-21 CFR 50;<br />

IRB regulations-21 CFR 56) for INDs and NDAs. In<br />

the presentation, Dr. Vuduthala listed requirements for<br />

a new IND with respect to chemistry/manufacturing/<br />

controls (CMC or Quality) data and pharmacologic and<br />

toxicologic data.<br />

On the subject of clinical trials, Dr. Vuduthala noted that<br />

the key to initiating clinical trials in the US is to ensure<br />

that a good safety and preclinical toxicology package is<br />

available for review in the initial IND stage along with<br />

a good clinical plan to evaluate proof of concept in<br />

healthy volunteers (Phase 1). Subsequently, performing<br />

dose ranging studies and confi rming effi cacy in a small<br />

population of relevant patients will need to be done in<br />

early Phase 2 studies. Based on the results, a detailed<br />

clinical plan is developed for Phase 2b and 3 pivotal<br />

clinical trials in a larger patient population. Throughout<br />

the drug development process, there are a number of<br />

opportunities for the industry to meet with members<br />

of the regulatory agency to discuss preclinical, clinical<br />

Balakumar, Jagadeesh: Pharmaceutical Discovery and Development<br />

and CMC plans and any issues requiring resolution.<br />

A pre-IND meeting, end of phase 2 meeting and pre-<br />

NDA meetings are keys to discuss the path forward<br />

and ensure the sponsor’s development plans are vetted<br />

by the Agency. Development of the product label<br />

(or package insert) occurs in parallel to the above, and<br />

labeling discussions are an important part of the NDA<br />

registration process.<br />

Biomarkers<br />

A fi nal topic discussed, which poses a great challenge<br />

to drug developers, regulators and physicians, is<br />

identifi cation of a ‘biomarker’, a biologic indicator,<br />

parameter or marker that can be used to measure<br />

the disease progress or the effects of treatment.<br />

The parameter can be physical or chemical as well as<br />

biological. A biomarker is defi ned as a characteristic<br />

that is objectively measured and evaluated as an<br />

indicator of normal biologic processes, pathogenic<br />

processes, or biological responses to a therapeutic<br />

intervention. 14 The last speaker of the symposium,<br />

Dr. Samba Reddy (Texas A&M University Health<br />

Science Center, USA) spoke on ‘Biomarkers in<br />

Preclinical and Clinical Drug Development’. Dr. Reddy<br />

focused on current knowledge on biomarkers in drug<br />

development (Box 4).<br />

Dr. Reddy discussed differences between diseaserelated<br />

and drug-related biomarkers. Disease-related<br />

biomarkers give an indication of the disease pathways<br />

or disease mechanism; whereas drug-related biomarkers<br />

indicate whether a drug will be effective in a specifi c<br />

patient and how the patient’s body will process it. The<br />

speaker covered the various aspects of biomarkers,<br />

including ‘Research Breaking in Classifi ed Biomarkers’,<br />

‘Biomarker Discovery Technology’, ‘Biomarker<br />

Quantifi cation and Assay’, ‘Biomarkers in Drug<br />

Discovery and Development’, ‘Disease Biomarkers<br />

and Healthcare’ and ‘Cancer Biomarkers’. The rapid<br />

emergence and evolution of translational medicine have<br />

Box 4: Traits of Biomarkers<br />

1. A biomarker can be a substance that is introduced into an<br />

organism to examine organ function or other aspects of<br />

health<br />

2. Biomarkers can be used in early diagnosis, disease<br />

prevention, drug treatment, drug response, drug target<br />

identifi cation, etc.<br />

3. Biomarkers represent key tools and parameters for the<br />

accelerated new drug discovery and clinical development<br />

4. Biomarkers can be classifi ed into three types: Natural<br />

history markers; Drug activity markers; and Surrogate<br />

markers<br />

5. In clinical trials, the biomarker serves as a surrogate<br />

endpoint for evaluating clinical benefi ts<br />

12 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


pushed biomarkers into the spotlight for successful drug<br />

development. He said that established pharmaceutical<br />

companies are integrating the biomarker strategies into<br />

their research product pipelines. In concluding remarks,<br />

Dr. Reddy said that the path from biomarker discovery<br />

to commercialization is fraught with numerous obstacles.<br />

Novel technologies are needed for biomarker discovery,<br />

and such approaches would be helpful in streamlining<br />

the drug development process by reducing time and<br />

costs, as well as minimizing drug attrition associated<br />

with safety issues.<br />

ACKNOWLEDGEMENTS<br />

We sincerely thank Drs. Sandhya Mandelkar (Head,<br />

Department of Pharmaceutical Candidate Optimization<br />

(PCO), Bristol-Myers Squibb India Ltd., Biocon BMS<br />

R&D Center, Bengaluru, India), K.S. Rao (Chief Scientifi c<br />

Offi cer, Rao Toxicology Foundation, Bengaluru, and<br />

Editor-in-Chief of Toxicology International), Gopi<br />

Vudathala (Associate Vice President, Global Regulatory<br />

Affairs-Interface CMC Sanofi -Aventis, New Jersey, USA)<br />

and Samba Reddy (Associate Professor, & Principal<br />

Investigator, Texas A&M University Health Science<br />

Center, College of Medicine, College Station, Texas,<br />

USA) for providing us their presentation materials<br />

from which this meeting report was prepared. We also<br />

express our gratitude to Dr. B.R. Jagashetty (Drugs<br />

Controller for the State of Karnataka, and Chairman<br />

of the Local Organizing Committee of 63rd IPC),<br />

Dr. N. Udupa (Convener, Scientifi c Services of IPCA),<br />

and Mr. S.M. Mudda (Chairman, Local Scientifi c<br />

Balakumar, Jagadeesh: Pharmaceutical Discovery and Development<br />

Committee of 63rd IPC) for their inspiration and<br />

support for this symposium.<br />

REFERENCES<br />

1. US FDA. Protecting America’s health through human drugs. http://<br />

www.fda.gov/Drugs/ResourcesForYou/Consumers/ucm143455.htm.<br />

Accessed 6 February 2012.<br />

2. Adams CP, Brantner VV. Spending on new drug development. Health<br />

Econ 2010;19:130–41.<br />

3. Terstappen GC, Reggiani A. In silico research in drug discovery. Trends<br />

Pharmacol Sci 2001;22:23–6.<br />

4. Kramer JA, Sagartz JE, Morris DL. The application of discovery toxicology<br />

and pathology towards the design of safer pharmaceutical lead<br />

candidates. Nat Rev Drug Discov 2007;6:636–49.<br />

5. Hopkins AL, Groom CR. The druggable genome. Nature Rev Drug<br />

Discov 2002;1:727–30.<br />

6. Yildirim MA, Goh KI, Cusick ME, Barabasi AL, Vidal M. Drug–target<br />

network. Nat Biotech 2007;25:1119–26.<br />

7. Imming P, Sinning C, Meyer A. Drugs, their targets and the nature and<br />

number of drug targets. Nature Rev Drug Discov 2006;5:821–34.<br />

8. Rask-Andersen M, Almén MS, Schiöth HB. Trends in the exploitation of<br />

novel drug targets. Nat Rev Drug Discov 2011;10:579–90.<br />

9. Lipinski CA. Drug-like properties and the causes of poor solubility and<br />

poor permeability. J Pharmacol Toxicol Method 2000;44:235–49.<br />

10. Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates?<br />

Nat Rev Drug Discov 2004;3:711–5.<br />

11. CFSAN Redbook 2000: Toxicological Principles for the Safety Assessment<br />

of Food Ingredients. Toxicity testing are listed in chapters III and<br />

IV of the book. http://www.fda.gov/Food/GuidanceComplianceRegulatoryInformation/GuidanceDocuments/FoodIngredientsandPackaging<br />

/Redbook/default.htm. Accessed 6 February 2012.<br />

12. OECD guideline for testing of chemicals, Section 4: e.g., acute toxicity,<br />

repeated dose toxicity, genetic toxicology, reproductive toxicology,<br />

etc. http://www.oecd-ilibrary.org/environment/oecd-guidelines-forthe-testing-of-chemicals-section-4-health-effects_20745788.<br />

(Home page:<br />

http://www.oecd.org/home/.) Accessed 6 February 2012.<br />

13. Environmental Protection Agency (EPA) guidelines for toxicity testing:<br />

http://www.epa.gov/ocspp/pubs/frs/publications/Test_Guidelines/<br />

series870.htm. Accessed 6 February 2012.<br />

14. Biomarkers Defi nitions Working Group. Biomarkers and surrogate<br />

endpoints: Preferred defi nitions and conceptual Framework. Clin<br />

Pharmacol Ther 2001;69:89–95.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 13


Nanobiotechnology: An overview of drug<br />

discovery, delivery and development<br />

Bhupinder Singh Sekhon<br />

PCTE Institute of Pharmacy, Jhande, Near Baddowal Cantt (Ludhiana)-142 021, Punjab, India<br />

ABSTRACT<br />

Nanobiotechnology is a new technology concerned specifi cally with the functionalism and modifi cation of<br />

chemical-physical structures on a biomolecular scale, and also is the application of nanotechnology to the life<br />

sciences. Nanotechnology for biotechnology and pharmaceutical applications has progressed from the concept<br />

stage to commercialization. Nanobiotechnology represents the future of medicine and healthcare. Various physical,<br />

chemical, electrical tools and methods used to investigate biological nanoobjects include optical tools, nanoforce<br />

and imaging, surface methods, mass spectrometry and microfl udics. Its application has an impact on diagnostics,<br />

drug delivery as well as drug discovery. Nanobiotechnology focuses on various areas such as nanobiotechnology<br />

and cancer, drug discovery and tools, and nanobiotechnology and medicine. Applications are emerging from<br />

all branches of nanobiotechnology in medicine and pharmacy. Several technologies including nanoparticles<br />

and nanodevices such as nanobiosensors and nanobiochips have been used to improve drug discovery and<br />

development. Some nanosubstances such as fullerenes and dendrimers/biodendrimers could be potential drugs<br />

for the future. Moreover, nanobiotechnology has the potential for combining drug design and drug delivery.<br />

However, limitations of the available nanoparticles still to be resolved for their application in the drug-discovery<br />

studies exist. The benefi ts of nanotechnology are enormous and so these benefi ts should be maximized while<br />

efforts are made to reduce the risks.<br />

Keywords: Nanobiotechnology, nanobiomaterials, nanobiostructures, nanocarriers<br />

INTRODUCTION<br />

Nanotechnology is a broader term applicable<br />

to small things in nanometer range (roughly<br />

in the 1−100 nm size regime in at least<br />

one dimension). Nanobiotechnology is a<br />

big word made up of three parts: NANO<br />

is really, really tiny, BIO is living things,<br />

and TECHNOLOGY is about tools.<br />

Nanobiotechnology is an emerging area<br />

of science which is concerned with the<br />

application of tools and processes in order<br />

to assemble devices for the study of objects<br />

in biological systems. Nanobiotechnology<br />

is that branch of nanotechnology that deals<br />

with biological and biochemical applications<br />

or uses. Nanobiotechnology often studies<br />

existing elements of living organisms and<br />

nature to fabricate new nano-devices.<br />

Generally, nanobiotechnology refers to<br />

the use of nanotechnology to further<br />

the goals of biotechnology. Relationship<br />

of nanotechnology, biotechnology and<br />

nanomedicine is shown in Figure 1.<br />

Further, nanobiotechnology (an integration<br />

of physical sciences, molecular engineering,<br />

biology, chemistry and biotechnology) has<br />

yielded healthcare discoveries that have<br />

been used for drug delivery and diagnostic<br />

purposes. 1<br />

In medicine and pharmacology, nanobiotechnology<br />

opens up new perspectives in<br />

analytics and therapy. Medical applications<br />

of nanobiotechnology resulted in appearance<br />

of new fi eld i.e. nanomedicine. An<br />

increasing use of nanobiotechnology by<br />

the pharmaceutical industries includes<br />

(i) drug delivery, and (ii) disease therapy.<br />

The potential topics in nanobitechnology<br />

Review Ar cle<br />

Received Date : 17-11-2011<br />

Revised Date : 04-01-2012<br />

Accepted Date : 17-02-2012<br />

DOI: 10.5530/rjps.2012.1.3<br />

Address for<br />

correspondence<br />

Bhupinder Singh Sekhon<br />

PCTE Institute of Pharmacy<br />

Jhande, Near Baddowal Cantt)<br />

(Ludhiana)-142 021<br />

Punjab, India<br />

E-mail: sekhon224@yahoo.co<br />

Mobile: 91-161-9876242299<br />

www.rjps.in<br />

14 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

Figure 1: Relationship of nanotechnology, biotechnology and nanomedicine.<br />

include molecular bioprobes, nanoparticles and nanobiosystems,<br />

nanobiomaterials, biomolecular assemblies<br />

and supra-biomolecules, nanobiosensors and nanobiochips,<br />

BioNEMS and nano-biofl uidics, nanobiophotonics,<br />

single-molecule detection and manipulation<br />

and molecular motors. Experts are of the opinion that<br />

nanobiotechnology has the potential to yield a scientifi<br />

c and industrial revolution, as envisioned by the<br />

numerous programs on nanotechnology/nanobiotechnology<br />

launched over the last decade by councils and<br />

governments worldwide.<br />

The potential uses and benefi ts of nanobiotechnology<br />

are enormous. 2 Nano-systems in biology, the most<br />

complex and highly functional nanoscale materials and<br />

machines have been invented by nature. Proteins and<br />

nucleic acids, and other naturally occurring molecules<br />

(polymers) regulate and control biological systems with<br />

incredible precision. Many nanotechnologists are in fact<br />

drawing inspiration from biology to device new materials<br />

and devices. Moreover, nanotechnology/nanobiotechnology<br />

is expected to rule tomorrow’s world.<br />

NANOBIOSTRUCTURES<br />

The use of biological principles is becoming widespread in<br />

the design of nanomaterials. Representative examples of<br />

nanobiostructures include DNA nanostructure, peptide<br />

structure and biomimetics. 3D-DNA nanostructures<br />

have emerged as promising tools for biology and materials<br />

science. In this regard, DNA cages, nanotubes, dendritic<br />

networks, and crystals with deliberate variation of their<br />

size, shape, persistence length, and porosities exhibited<br />

dynamic character, allowing their selective switching<br />

with external stimuli. Short peptides can spontaneously<br />

associate to form nanotubes, nanospheres, nanofi brils,<br />

nanotapes, and other ordered structures at the nanoscale.<br />

Further, peptides can also form macroscopic assemblies<br />

such as hydrogels with nanoscale order. The theory and<br />

the mechanisms behind peptide self-assembly process<br />

and their bionanotechnology applications have been<br />

reported. 3 In general, nanobiotechnology requires<br />

the organization of atoms and molecules in a two- or<br />

three-dimensional space. Self-assembly properties of<br />

biomolecules have ability to spontaneously organize<br />

into nanostructures, which allows mimicking the living<br />

cell membranes.<br />

Nanoparticle usually forms the core of nanobiomaterial.<br />

Nanobiomaterial is made of nanoparticles and<br />

nanobiomaterials are emerging as the most hopeful<br />

area of research within the area of biological materials<br />

science and engineering. They have an increased number<br />

of atoms and crystal grains at their surfaces and possess<br />

a higher surface area to volume ratio than conventional<br />

microscale biomaterials. These differences in surface<br />

topography alter the corresponding surface energy<br />

for protein adsorption. Nanobiomaterials can be used<br />

for human implant, orthopedics, drug delivery, gene<br />

therapy, antimicrobial treatments, array technologies,<br />

and diagnostics. Moreover, nanobiomaterials help<br />

with targeting, measuring, sensing, and imaging. The<br />

approaches used in constructing nanobiomaterials are<br />

given in Figure 2.<br />

NANOBIOTECHNOLOGY BASED DRUG DELIVERY/<br />

DEVELOPMENT<br />

Nanomedical approaches to drug delivery center<br />

on developing nanoscale particles or molecules to<br />

improve the bioavailability of a drug. Nanomaterials<br />

and nanoparticles are likely to be cornerstone of<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 15


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

Figure 2: Specific configuration of nanobiomaterial when applied to biological or medical applications.<br />

innovative nanomedical devices employed for drug<br />

discovery and delivery, discovery of biomarkers<br />

and molecular diagnostics. 4 Preparation methodsproperties<br />

of nanoscale systems including liposomes,<br />

micelles, emulsions, nanoparticulates, and dendrimer<br />

nanocomposites, and clinical indications are known<br />

for imaging in vivo, sustained and targeted delivery of<br />

drugs, genes, and proteins. The recent development<br />

in drug delivery system has opened up new potential<br />

& possibility for production of pharmaceutical drugs,<br />

oral peptides, gene therapy and nanocosmeceuticals. 5<br />

Nanobiotechnology exploits nanotechnology and<br />

biotechnology to analyze and create nanobiosystems<br />

to meet a wide variety of challenges and develops a<br />

wide range of applications. Biomaterials like DNA and<br />

proteins combined with electronic systems consequently<br />

results in the formation of new devices, sensors, and<br />

systems. Examples include (i) a carbon nanotube<br />

with single-stranded DNA wrapping around it. (ii) a<br />

graphene sheet with duplex DNA molecules. (iii) a<br />

nanoparticle attachment with antibodies, (iv) a nanorod<br />

after enzyme immobilization and (v) Streptavidin<br />

(protein) attachment to nanoparticles along with biotin<br />

(protein). 6 Several technologies, including nanoparticles<br />

and nanodevices such as nanobiosensors, nanobiochips,<br />

nanosubstances were used to improve drug discovery<br />

and development. 7<br />

Gold nano rod-DARPP-32siRNA complexes (nanoplexes)<br />

have been reported suitable for brain-specifi c<br />

delivery of appropriate siRNA for therapy of drug<br />

addiction and other brain diseases. 8 Boron nitride nanotubes<br />

are structural analogues of carbon nanotubes in<br />

nature: alternating B and N atoms entirely substitute for<br />

C atoms in a graphitic like sheet with almost no change<br />

in atomic spacing. By virtue of their magnetic properties,<br />

boron nitride nanotubes could be exploited for<br />

magnetic, physically guided, drug targeting. The interactions<br />

between boron nitride nanotubes and living cells<br />

were reported and the piezoelectric properties of boron<br />

nitride nanotubes make them attractive candidates as<br />

bionanotransducers for cell sensing and stimulation,<br />

a use which still has to be exploited. 9<br />

DENDRIWORMS<br />

Dendriworms (magnetic nanoworm + dendrimer) are<br />

synthetic polymers that can be used to carry a large range<br />

of molecules such as siRNA (made up of magnetic<br />

nanoparticles as well as a fl uorescent nanoparticle)<br />

which allow the nanoworm to be traced as to where<br />

it is. Tremendous progress has been achieved in the<br />

recent years in our understanding of the ability of small<br />

interfering RNAs to silence gene expression in mammalian<br />

cells. This has provided us with a revolutionary new tool<br />

to modulate the expression of disease-causing genes.<br />

Short interfering RNAs (siRNAs) have emerged as a<br />

potent new class of therapeutics, which regulate gene<br />

expression through sequence-specifi c inhibition of<br />

mRNA translation. Lipid-, polymer-, and nanoparticlebased<br />

siRNA delivery vehicles have proven effective<br />

in improving the stability, bioavailability, and target<br />

specifi city of siRNAs following systemic administration<br />

in vivo. Additionally, these methods provided a platform<br />

to modify siRNAs with a variety of contrast agents and<br />

16 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

have enabled nuclear and magnetic resonance imaging<br />

of siRNA delivery in preclinical studies. Such imageguided<br />

delivery approaches represent a crucial step in<br />

the transition of siRNA therapeutics to the clinic. 10<br />

Scientists reported the development of dendriworms<br />

as a modular platform for siRNA delivery in vivo.<br />

Researchers have demonstrated that siRNA-carrying<br />

dendriworms can be readily internalized by cells and<br />

enabled endosomal escape across a wide range of loading<br />

doses, whereas dendrimers or nanoworms alone were<br />

ineffi cient. In addition, dendriworms carrying siRNA<br />

against the epidermal growth factor receptor reduced<br />

protein levels of epidermal growth factor receptor in<br />

human glioblastoma cells by 70−80%, 2.5-fold more<br />

effi ciently than commercial cationic lipids. Reported<br />

data established dendriworms as a multimodal platform<br />

that enabled fl uorescent tracking of siRNA delivery<br />

in vivo, cellular entry, endosomal escape, and knockdown<br />

of target proteins. 11<br />

INORGANIC NANOCRYSTALS<br />

Researchers reported the fi rst use of inorganic<br />

fl uorescent lanthanide (europium and terbium) ortho<br />

phosphate [LnPO ·H O, Ln = Eu and Tb] nanorods as<br />

4 2<br />

a novel fl uorescent label in cell biology. These nanorods,<br />

synthesized by the microwave technique, retained their<br />

fl uorescent properties after internalization into human<br />

umbilical vein endothelial cells (HUVEC), 786-O cells,<br />

or renal carcinoma cells (RCC). At concentrations up to<br />

50 μg/ml, the use of [ 3H]-thymidine incorporation assays,<br />

apoptosis assays (TUNEL), and trypan blue exclusion<br />

illustrated the non-toxic nature of these nanorods,<br />

a major advantage over traditional organic dyes. 12<br />

DESIGN OF NEW-AGE DELIVERY SYSTEMS<br />

Scientists have shown a modifi ed version of the bacteriophage<br />

phi29 DNA-packaging motor, when reconstituted<br />

into liposomes and inserted into planar lipid bilayers,<br />

allowed the translocation of double-stranded DNA.<br />

Moreover, this engineered and membrane-adapted<br />

phage connector is expected to have applications in<br />

microelectromechanical sensing, microreactors, gene<br />

delivery, drug loading and DNA sequencing. 13<br />

Aptamers<br />

Aptamers, single stranded DNA or RNA molecules,<br />

generated by a method called SELEX (systematic<br />

evolution of ligands by exponential enrichment) have<br />

been widely used in various biomedical applications.<br />

Technology combination of nanobiotechnology<br />

with aptamers opened the way to more sophisticated<br />

applications in molecular diagnosis. Recent developments<br />

in SELEX technologies and new applications of<br />

aptamers have the ability to discriminate between two<br />

closely related targets, for example a cancerous cell and<br />

an untransformed cell of the same tissue type, makes<br />

aptamers suitable as imaging reagents for non-invasive<br />

diagnostic procedures. 14<br />

Implantable biomedical devices<br />

A bilayer structure comprising a thin gold layer and<br />

a polypyrrole fi lm has been developed as a valve and<br />

holds promise for implantable biomedical devices. 15<br />

Implantable medical devices that comprise a substrate<br />

and a porous layer comprising close packed spherical<br />

pores disposed over the substrate have been reported<br />

and the porous layer may also comprise a therapeutic<br />

agent. 16<br />

Nanoprobes<br />

Tiny nanoprobes have shown to be effective in delivering<br />

cancer drugs more directly to tumor cells - mitigating<br />

the damage to nearby healthy cells - and research has<br />

shown that the nanoprobes are getting the drugs to<br />

right cellular compartments. 17 Carbon nanotubes’<br />

unique properties including low cytotoxicity and good<br />

biocompatibility attract their use as vector system in<br />

target delivery of drugs, proteins and genes. 18<br />

Researchers have focused on the integration of biological<br />

molecules (DNA, antibodies, and enzymes)<br />

into micro- and nanostructures, with state-of-theart<br />

bioelectronic read-out systems, extracting useful<br />

analytical signals with interest for various fi elds. In<br />

this direction, nanotechnology based biosensors are<br />

the product of this integration with great interest for<br />

several applications that aim at a signifi cant improve<br />

of the quality and security of citizen’s life. 19 Rapamune<br />

was the fi rst product based on approved NanoCrystal<br />

Technology. Nanodrugs and nanodevices approved by<br />

the FDA, and emerging nanoproducts that may pose<br />

a challenge for current regulatory schemes both in the<br />

U.S. and internationally, have been reported. 20<br />

Scientists have designed a novel drug delivery vehicle<br />

by hybridizing macrophages with nanoparticles through<br />

cell surface modifi cation. Nanoparticles immobilized<br />

on the cell surface provided numerous new sites for<br />

anticancer drug loading, hence potentially minimizing<br />

the toxic effect of anticancer drugs on the viability and<br />

hypoxia-targeting ability of the macrophage vehicles.<br />

In particular, quantum dots and 5-(aminoacetamido)<br />

fl uoresceinlabeled polyamidoamine dendrimer G4.5,<br />

both of which were coated with amine-derivatized<br />

polyethylene glycol, were immobilized to the sodium<br />

periodate-treated surface of RAW264.7 macrophages<br />

through a transient Schiff base linkage. Further, a<br />

reducing agent, sodium cyanoborohydride, was applied<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 17


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

to reduce Schiff bases to stable secondary amine linkages.<br />

The distribution of nanoparticles on the cell surface was<br />

confi rmed by fl uorescence imaging, and it was found to<br />

be dependent on the stability of the linkages coupling<br />

nanoparticles to the cell surface. 21<br />

Protein therapy<br />

Protein therapy, which delivers proteins into the cell<br />

to replace the dysfunctional protein, is considered the<br />

most direct and safe approach for treating disease and<br />

has been looked into as a possible alternative to gene<br />

therapy. Scientists have demonstrated a general, effective;<br />

low-toxicity intracellular protein delivery system based<br />

on single-protein nanocapsules, and opened a new<br />

direction not only for protein therapy but also for<br />

cellular imaging, tumor tracking, cosmetics and many<br />

other applications. 22 RNAi screen of the protein kinome<br />

identifi ed checkpoint kinase 1 (CHK1) as a therapeutic<br />

target in neuroblastoma. 23<br />

Stabilizing proteins at high concentration is of broad<br />

interest in drug delivery, for treatment of cancer<br />

and many other diseases. Scientists created highly<br />

concentrated antibody dispersions (up to 260 mg/ml)<br />

comprising of dense equilibrium nanoclusters of<br />

protein [monoclonal antibody (mAb) 1B7, polyclonal<br />

sheep Immunoglobulin G (IgG) and bovine serum<br />

albumin (BSA)] molecules, which upon dilution in vitro or<br />

administration in vivo, remained conformationally stable<br />

and biologically active. The nanoclusters are formed by<br />

adding trehalose as a co-solute which strengthened the<br />

short-ranged attraction between protein molecules. The<br />

protein cluster diameter was reversibly tuned from 50 to<br />

300 nm by balancing short-ranged attraction against<br />

long–ranged electrostatic repulsion of weakly charged<br />

protein at a pH near the isoelectric point (pI). 24<br />

Dendrimers/biodendrimers<br />

Dendrimers are nanostructures produced from<br />

macromolecules such as polyamidoamine, polypropyleneimine<br />

and polyaryl ether; and are highly branched with<br />

an inner core. The particle size range is between 1 to<br />

100 nm although their sizes are mostly less than 10 nm.<br />

Biodendrimers are dendrimers composed of repeating<br />

units known to be biocompatible or biodegradable<br />

in vivo to natural metabolites. Medical applications of<br />

dendrimers as multifunctional nanosized containers are<br />

shown in Figure 3.<br />

Dendrimers have successfully proved themselves<br />

as useful additives in different routes of drug<br />

administration to be applied in routes with particular<br />

reference to intravenous, oral, transdermal, and ocular<br />

delivery systems. 25 Dendrimers can act as vectors, in<br />

gene therapy and as an agent for Photodynamic Therapy<br />

of tumorigenic keratinocytes. 26<br />

Figure 3: Some medical applications of dendrimers/biodendrimers.<br />

NANOSTRUCTURED DRUG DELIVERY SYSTEMS<br />

Bionanofabrication<br />

2D crystallization of proteins, especially engineered<br />

proteins, is emerging as a powerful tool for bottom-up<br />

approaches to the nanofabrication of functional<br />

structures. Some aspects of 2D protein crystallization,<br />

including key approaches to growing 2D protein crystals<br />

and their potential applications ranging from biosensors,<br />

diagnostic kits, vaccine applications, and templates for<br />

mineral formation have been reported. 27<br />

Nanostructured lipid carriers (NLC) that can deliver<br />

active pharmaceutical ingredients across the skin have<br />

emerged as novel systems composed of physiological<br />

lipid materials suitable for topical, dermal and<br />

transdermal administration. The design characteristics,<br />

production and composition of semi-solid formulations<br />

containing NLC as API carriers (for example hydrogels)<br />

have been reported. 28 Results of characterization studies<br />

strongly supported the potential application of these<br />

drugs-loaded NLC as prolonged delivery systems for<br />

lipophilic drugs by several administration routes, in<br />

particular for intravenous administration. 29<br />

Fullerenes for Medical Diagnostics<br />

Scientists have created new materials from Fullerenes by<br />

fi lling them with atoms of various metals. An important<br />

example is a fullerene that encases a sensitive contrast<br />

agent (gadolinium) for MRI applications, including as<br />

a diagnostic and therapeutic agent for brain tumors.<br />

Researchers have co-invented a hands-off process for<br />

fi lling fullerenes with radioactive material and these fi nding<br />

could be utilized in medical applications, such as MRIs<br />

and diagnostic and therapeutic agents for brain tumors. 30<br />

NANOBIO TECHNOLOGY AND TISSUE ENGINEERING<br />

Scientists discussed the current applications of nanoscale<br />

materials to bladder tissue engineering. 31 Researchers<br />

18 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

explored the bottom-up and top-down nanofabrication<br />

technologies and their use in various drug delivery and<br />

tissue engineering applications. 32<br />

NANOBODIES<br />

The structural properties of Nanobodies and their<br />

possible therapeutic applications were discussed and<br />

data from phase I clinical trials of the novel ‘fi rstin-class’<br />

anti-thrombotic agent ALX-0081 (Ablynx NV)<br />

were reported. 33 Results suggested that the generation<br />

of Nanobody-displaying immune phage libraries and<br />

subsequent in vivo biopanning in appropriate animal<br />

models is a promising approach for the identifi cation of<br />

novel vascular targeting agents. 34<br />

NANOBOTS/NANOROBOTS<br />

Nanorobots could be employed for the diagnostics,<br />

targeted drug dispensation, elimination of xenogenous<br />

particles from the body, and repair of cells and tissues,<br />

e.g. the skin and teeth. 35 Nanorobots could also be<br />

programmed to perform delicate surgeries or remove<br />

obstructions in the circulatory system. 36<br />

BIOMARKERS<br />

Biomarkers are molecules that can be measured in blood,<br />

other body fl uids, and tissues to assess the presence or<br />

state of a disease. They have the potential to help us<br />

detect cancer earlier, determine a tumor’s aggressiveness,<br />

or predict a patient’s response to a particular treatment.<br />

Biomarkers play a role in use of pharmacogenetics,<br />

pharmacogenomics and pharmacoproteomics for<br />

development of personalized medicine. Label-free<br />

nanosensors can detect disease markers to provide<br />

point-of-care diagnosis that is low-cost, rapid, specifi c<br />

and sensitive. Scientists showed specifi c and quantitative<br />

detection of two model cancer antigens from a 10 μl<br />

sample of whole blood in less than 20 min. They used<br />

nanowire sensors to detect and measure concentrations<br />

of two specifi c biomarkers: one for prostate cancer<br />

and the other for breast cancer. This novel device acted<br />

as a fi lter, catching the biomarkers, antigens specifi c<br />

to prostate and breast cancer on a chip while washing<br />

away the rest of the blood. 37 The emerging trends in<br />

the development of biomarkers for early detection and<br />

precise evaluation of cancer disease were reported. 38<br />

This bio-nano-info convergence holds great promise for<br />

molecular diagnosis and individualized therapy of cancer<br />

and other human diseases. Scientists have demonstrated<br />

the feasibility of multiplex detection using the surfaceenhanced<br />

Raman scattering-based molecular sentinel<br />

(MS) technology in a homogeneous solution. Two MS<br />

nanoprobes tagged with different Raman labels were<br />

used to detect the presence of the erbB-2 and ki-67<br />

breast cancer biomarkers. The multiplexing capability of<br />

the MS technique was demonstrated by mixing the two<br />

MS nanoprobes and tested in the presence of single or<br />

multiple DNA targets. 39<br />

NANOBIOTECHNOLOGY IN CANCER<br />

Application of nanotechnology/nanobiotechnology in<br />

biomedicine may contribute to signifi cant advances in<br />

imaging diagnosis and treatment of cancer. Quantum<br />

dots, gold nanoparticles, magnetic nanoparticles,<br />

carbon nanotubes, gold nanowires and many other<br />

materials have been developed over the years, alongside<br />

the discovery of a wide range of biomarkers to lower<br />

the detection limit of cancer biomarkers. Current<br />

developments in cancer detection methods with an<br />

emphasis on nanotechnology were reported. 40 Some<br />

of the recent development in nanotechnologies and<br />

their applications in diagnosing and developing cancer<br />

therapies have been reported. 41<br />

Scientists reported a cooperative nanosystem consisting<br />

of two discrete nanomaterials that work in concert within<br />

the bloodstream to locate, adhere to and kill cancerous<br />

tumors. While one type of nanoparticle improved<br />

detection of the tumor, the other was designed to kill the<br />

tumor. The fi rst component consists of gold nanorod<br />

“activators” that populate the porous tumor vessels and<br />

act as photothermal antennas to specify tumor heating<br />

via remote near-infrared laser irradiation. In addition,<br />

it was found that local tumor heating accelerated the<br />

recruitment of the second component: a targeted<br />

nanoparticle consisting of either magnetic nanoworms<br />

(NW) or doxorubicin-loaded liposomes (LP). Nineamino<br />

acid peptide LyP-1 (Cys-Gly-Asn-Lys-Arg-Thr-<br />

Arg-Gly-Cys) was employed as the targeting species that<br />

binds to the stress-related protein, p32, which was up<br />

regulated on the surface of tumor-associated cells upon<br />

thermal treatment. Mice containing xenografted MDA-<br />

MB-435 tumors that were treated with the combined<br />

gold nanorod /LyP-1- doxorubicin-loaded liposomes<br />

therapeutic system displayed signifi cant reductions in<br />

tumor volume compared with individual nanoparticles<br />

or untargeted cooperative system. 42<br />

A new nanoparticle formulation can be magnetically<br />

guided to deliver and silence genes in cells and tumors<br />

in mice. This formulation, termed LipoMag, consisted<br />

of an oleic acid-coated magnetic nanocrystal core<br />

and a cationic lipid shell. When compared with the<br />

commercially available PolyMag formulation, LipoMag<br />

displayed more effi cient gene silencing in 9 of 13 cell<br />

lines, and better anti-tumour effects when systemically<br />

administered to mice bearing gastric tumours. By<br />

delivering an optimized sequence of a silencing RNA<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 19


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

that targets the epidermal growth factor receptor of<br />

tumor vessels, the intended therapeutic benefi t was<br />

achieved with no evident adverse immune reaction or<br />

untoward side effects. 43<br />

NANOMEDICINE<br />

Nanomedicine has triggered the wind of revolution in<br />

medicine and important areas that nanomedicine covers<br />

include targeted drug delivery in cancer treatment,<br />

biotechnology, disease diagnosis to nanostructure<br />

implants. 44<br />

A variety of recent research combines Heparin (HP) and<br />

nanomaterials for a myriad of applications. HP has been<br />

conjugated to the surface of the nanoparticles, such as<br />

magnetic and metallic nanoparticles, or biodegradable<br />

and nondegradable synthetic polymers. HP has also<br />

been incorporated into the nanoparticles. The different<br />

possibilities of HP-based nanoparticle composites and<br />

their medicinal or biological applications were reported. 45<br />

It was qualitatively demonstrated that enhanced<br />

fl uorescence emission signals occurred from clustered<br />

QDs and deduced that the band 3 membrane proteins<br />

in erythrocytes were clustered. 46<br />

Nanonephrology<br />

Nanonephrology is a branch of nanomedicine and<br />

nanotechnology that deals with (i) the study of kidney<br />

protein structures at the atomic level (ii) nano-imaging<br />

approaches to study cellular processes in kidney cells and<br />

(iii) nanomedical treatments that utilize nanoparticles<br />

and to treat various kidney diseases. Various devices<br />

based on nanotechnology are used for the studying<br />

the different kidney processes and detecting disorders.<br />

Nanotechnological fi lters have the potential to provide<br />

immediate relief for dialysis patients. 47 Scientists have<br />

demonstrated a new artifi cial renal chip by integrating<br />

a high effi cient biocompatible polymeric nanofi bers<br />

membrane with the polydimethylsiloxane(PDMS)<br />

based micro-fl uidic platform via the optimization of<br />

PDMS micro fl uidic channel network and the multiple<br />

packaging of nanofi bers membrane. 48<br />

Nanobiopharmaceutics<br />

Nanobiopharmaceutics aims at the development of<br />

innovative multidisciplinary approaches for the design,<br />

synthesis and evaluation of molecular, nano- and microscale<br />

functionalities for targeted delivery of therapeutic<br />

peptides and proteins (biopharmaceutics). Nanoscale<br />

biomaterials are categorized by metal, non-metal, carbon,<br />

polymer, lipid, virus and miscellaneous nanostructures<br />

as nanobiopharmaceutical carrier systems and their<br />

medical/biological applications as well as toxicological<br />

issues in the fi eld of biomedical nanotechnology<br />

were reported. 49 The basis of technologic application<br />

in biopharmaceutics is Nanoscale Drug Carrying<br />

System and the various units of the system consist of<br />

liposome system which varies between 30 nm to several<br />

micrometers in diameter with their characteristics<br />

governed by their size, surface composition and charge.<br />

Surface modifi ed liposomes carrying doxorubicin and<br />

antisense oligonucleotide system have successfully<br />

targeted multidrug resistance associated protein,<br />

messenger RNA and bcl2 RNA. System after reaching<br />

the cell, delivered the doxorubicin and the antisense<br />

oligonucleotides successfully and inhibited the synthesis<br />

of MRP1 and bc12 RNA and provoked the apoptosis<br />

of carcinomatous cell by arousing the caspase (cysteineaspartic<br />

proteases) dependent pathway. 50<br />

A “nanoviricide” is a fl exible nano-scale material<br />

approximately a few billionths of a meter in size, which<br />

is chemically programmed to specifi cally target and<br />

attack a particular type of virus like a guided missile.<br />

NV-INF-1, the selected candidate drug substance of the<br />

FluCide program, has been shown to be highly effective<br />

in controlling infl uenza viral infection in lethal infection<br />

mouse model. NanoViricides nanotechnology possesses<br />

potent antiviral effi cacy by targeting the mechanisms by<br />

which viruses attach or bind to cells. 51<br />

Nanopharmacology<br />

Nanopharmacology is the use of nanotechnology<br />

for (i) discovery of new pharmacological molecular<br />

entities; (ii) selection of pharmaceuticals for specifi c<br />

individuals to maximize effectiveness and minimize side<br />

effects; and (iii) delivery of pharmaceuticals to targeted<br />

locations or tissues within the body . The potential<br />

applications of biochips, nanosensors, bioreactors,<br />

neural stem cells, immune nanoparticles, biodegradable<br />

polymers, and convection-enhanced drug delivery in the<br />

diagnostics and treatment of diseases were reported.<br />

Numerous novel medicinal forms were reported,<br />

including polymeric nanoparticles, nanotubes, micelles,<br />

liposomes, dendrimers, fullerenes, and hydrogels. In<br />

particular, highly stable glycosphingolipid nanotubes<br />

and nanoliposomes were proposed as drug delivery<br />

systems. For this purpose, the model of stimulation of<br />

skin vasomotor reactions by nitroglycerin application<br />

was developed. The effect of nitroglycerin was found to<br />

increase 1.5 times when used in the form of dispersion<br />

with nanotubes as carriers and almost 2.5 times in the<br />

case of dispersion with nanoliposomes as carriers.<br />

Nanotechnologically manufactured biologically active<br />

substances Apiton-25 (containing apis products) and<br />

Microhydrin (containing SiO nanoparticles, silicon<br />

2<br />

hydrogen bonds Si-H, and free negative charges for free<br />

radical neutralization) results have indicated that both<br />

Apiton-25 and Microhydrin upon peroral administration<br />

20 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

enhanced the cyclic trial performance during prolonged<br />

submaximal exercise in endurance-trained cyclists. 52<br />

Scientists have developed a combination drug that<br />

promises a safer, more precise way for medics and fellow<br />

soldiers in battle to give a fallen soldier both morphine<br />

and a drug that limits morphine’s dangerous side effects.<br />

They have used nanotechnology to devise ultra-small<br />

polymer particles capable of carrying the drugs into<br />

the body. The development of the combination drug<br />

makes possible a precise feedback system that can safely<br />

regulate release of the drugs aboard the nanoparticles. 53<br />

NANOBIOTECHNOLOGY AND PERSONALIZED<br />

MEDICINE<br />

Nanobiotechnology will facilitate the integration<br />

of diagnostics with therapeutics and facilitate the<br />

development of personalized medicine. 54 Nanobiotechnology<br />

is being used to refi ne discovery of biomarkers,<br />

molecular diagnostics, drug discovery and<br />

drug delivery, which are important basic components of<br />

personalized medicine. 55 Personalized management of<br />

cancer, facilitated by nanobiotechnology, is expected to<br />

enable early detection of cancer, more effective and less<br />

toxic treatment increasing the chances of cure. 56<br />

Nanobiomaterials/Nanobioanalysis/<br />

Nanobiochips<br />

Novel opportunities and challenges offered by nanobiomaterials<br />

in tissue engineering have been reported. 57<br />

The current state of development of nanodiagnostic<br />

technologies including nanobiochips and nanobiosensors<br />

has been reported. 58 Neuronanobiotechnology<br />

based delivery system are developing rapidly and one<br />

example of current nanobiotechnological research<br />

involved nanospheres coated with fl uorescent polymers.<br />

59 An assay based on gold nanoparticles could<br />

detect recurrences of prostate cancer sooner than is<br />

possible with existing techniques. 60 Carbon nanotubes<br />

have been used to probe the properties of bilayer systems<br />

resembling living cell membranes. 61<br />

A multifunctional one-dimensional nanostructure<br />

incorporating both CdSe quantum dots (QDs) and Fe O 3 4<br />

nanoparticles (NPs) within a SiO -nanotube matrix was<br />

2<br />

successfully synthesized based on the self-assembly of<br />

preformed functional NPs, allowing for control over the<br />

size and amount of NPs contained within the composite<br />

nanostructures. This specifi c nanostructure is distinctive<br />

because both the favorable photoluminescent and<br />

magnetic properties of QD and NP building blocks<br />

were incorporated and retained within the fi nal silicabased<br />

composite, thus rendering it susceptible to both<br />

magnetic guidance and optical tracking. Moreover, the<br />

resulting hydrophilic nanocomposites were found to<br />

easily enter into the interiors of HeLa cells without<br />

damage, thereby highlighting their capability not<br />

only as fl uorescent probes but also as possible drugdelivery<br />

vehicles of interest in nanobiotechnology. 62<br />

A large variety of nanobioanalysis methods have been<br />

reported. 63 Scientists have described the construction<br />

and use of two major classes of nano-bio-chip designs<br />

that serve as cellular and chemical processing units.<br />

These nanobiochips possess capabilities for measuring<br />

such diverse analyte classes as cells, proteins, DNA and<br />

small molecules in the same compact device. Further,<br />

applications such as disease diagnosis and prognosis<br />

for areas including cancer, heart disease and HIV were<br />

reported. 64 One-dimensional nanostructures such as<br />

nanowires are ideal for diagnosis as they can be integrated<br />

into microfl uidic chips that provide a complete sensor<br />

system. 65<br />

DNA nanomachines are synthetic assemblies that<br />

switch between defi ned molecular conformations<br />

upon stimulation by external triggers. Researchers have<br />

reported the construction of a DNA nanomachine called<br />

the I-switch, which is triggered by protons and functions<br />

as a pH sensor based on fl uorescence resonance energy<br />

transfer inside living cells. Moreover, this was found<br />

an effi cient reporter of pH from pH 5.5 to 6.8, with<br />

a high dynamic range between pH 5.8 and 7, thereby,<br />

illustrating the potential of DNA scaffolds responsive<br />

to more complex triggers in sensing, diagnostics and<br />

targeted therapies in living systems. 66<br />

INTEGRATION OF RECOMBINANT TECHNOLOGY<br />

AND NANOSCIENCE<br />

Recombinant technology is the most important<br />

prerequisite for the effective engineering of nanostructured<br />

deoxyribonucleic acid and protein based<br />

materials in nanoscience. This technology allowed the<br />

manipulation of the properties of molecules, including<br />

physico-chemical properties of proteins that control<br />

electron transport and photochemical processes in<br />

the development of molecular electronic devices and<br />

device fabrication. Recombinant molecules, such as<br />

recombinant ovalbumin and recombinant ovalbumin<br />

mutants have provided a powerful means for the study<br />

of their physico-chemical and structural characteristics,<br />

and thereby for their use in nanoscience. The researchers<br />

have provided an overview of the integration of<br />

recombinant technology and nanoscience through<br />

reported studies in areas, including food, environment,<br />

medicine, physics and chemistry. 67<br />

CONCLUSIONS AND PERSPECTIVES<br />

Nanobiotechnology is an emerging fi eld that seeks new<br />

solutions to pressing health and environmental problems<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 21


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

by combining physical sciences and engineering with<br />

life sciences and medicine. This exciting frontier of<br />

discovery is generating new therapies, devices, diagnostic<br />

tools, and a better understanding of the relationship<br />

between cells and disease. Nanobiotechnology also<br />

deals with the investigation and utilization of the newly<br />

conceived nanomaterials, as well as the construction of<br />

functionalized nanobiosystems. Nanopharmaceuticals<br />

offer the ability to detect diseases at much earlier<br />

stages. Drug delivery is the most advanced tool of<br />

nanobiotechnology. Nanomedicine combines nanotechnology<br />

and medicine, and nanotherapeutics is the<br />

use of nanomedicine in therapy. The ultimate goal of<br />

nanotherapeutic is comprehensive monitoring repair<br />

and improvement of all human biologic system.<br />

Nanobiotechnology should provide many of the tools<br />

necessary to enable the components that will one day be<br />

inserted into commercialized products.<br />

Genomics, proteomics, and metabolomics combined<br />

with the power of nanobiotechnology have the potential<br />

to understand the disease in a way that was previously<br />

not possible and it is expected that the disease will<br />

be targeted more effectively and precisely. Experts<br />

support a comprehensive technology assessment of<br />

nanobiotechnologies along with a full-blown risk/benefi t<br />

analysis before their adoption. New biofriendly imaging<br />

probes based on functionalized inorganic nanocrystals are<br />

being developed and are available to facilitate state-of-theart<br />

bioimaging studies. The research on the combination<br />

of chemotherapeutic agents with gene therapy should<br />

be further strengthened to overcome the limitations of<br />

conventional cancer treatment. In this context, a strongly<br />

synergistic antiproliferative effect was observed in colon<br />

cancer cells when E gene expression was combined with<br />

the activity of the 5-Fluorouracil–loaded biodegradable<br />

poly(ε-caprolactone) nanoparticles, thereby indicating<br />

the potential therapeutic value of the combined<br />

therapy. 68 It is hoped that nanobiotechnology will extend<br />

the limits of current molecular diagnostics and enable<br />

point of care diagnostics, integration of diagnostics<br />

with therapeutics, and development of personalized<br />

medicine. Three-dimensional nanobiostructure-based<br />

self-contained devices consisting of a glucose/oxygen<br />

sensor, a biofuel cell and a wireless signal transmitter to<br />

demonstrate wireless monitoring of glucose and oxygen<br />

in biological fl uids, wounds, and cell cultures is under<br />

development. 69 As yet there are no directives to regulate<br />

nanobiotechnology by various regulating bodies but as<br />

products are ready to enter market, these are expected<br />

to be in place. The largest expansion is expected in<br />

coming years. 70 Nanobiotechnology is likely to trigger<br />

advances in the early detection of a variety of diseases<br />

and improvements in biological implants. Experts are of<br />

the opinion that efforts should be made to educate, and<br />

increase awareness about nanobiotechnology through a<br />

transparent public dialogue.<br />

REFERENCES<br />

1. Nalwa HS. (Ed.). Handbook of nanostructured biomaterials and<br />

their applications in nanobiotechnology, Volume 2: Applications in<br />

nanobiotechnology. American Scientifi c Publishers, 2005.<br />

2. Jain KK. The role of nanobiotechnology in drug discovery. In:<br />

Pharmaceutical Biotechnology-Volume 655 of Advances in experimental<br />

medicine and biology, Edited by: Carlos A. Guzmán and Giora Z.<br />

Feuerstein, Chapter 4, Springer, 2009. pp 37–43.<br />

3. Castillo-León J, Sasso L, Svendsen W. Self-assembled peptide<br />

nanostructures: Advances and applications in nanobiotechnology, CRC<br />

Press, Taylor and Francis Groups, 2012.<br />

4. Jain KK (ed). The Handbook of biomarkers. Series: Neuromethods,<br />

Vol. 45, Springer, 2010.<br />

5. Irvine DJ. Drug delivery: One nanoparticle, one kill. Nat. Mater. 2011;10:<br />

342–3.<br />

6. Amin R, Hwang S, Park SH. Nanobiotechnology: an interface between<br />

nanotechnology and biotechnology. NANO: Brief Reports and Reviews<br />

2011;6( 2):101–11.<br />

7. Jain KK. The role of nanobiotechnology in drug discovery. Drug Discov<br />

Today 2005;10:1435–42.<br />

8. Bonoiu AC, Mahajan SD, Ding H, et al. Nanotechnology approach for<br />

drug addiction therapy: gene silencing using delivery of gold nanorodsiRNA<br />

nanoplex in dopaminergic neurons. Proc Natl Acad Sci U S A.<br />

2009;106:5546–50.<br />

9. Ciofani G, Raffa V, Menciassi A, Cuschieri A. Boron nitride nanotubes:<br />

an innovative tool for nanomedicine. Nanotoday 2009;4(1):8–10.<br />

10. Ifediba MA, Moore A. In vivo imaging of the systemic delivery of small<br />

interfering RNA. Wiley Interdisciplinary Reviews: Nanomedicine and<br />

Nanobiotechnology 2012:1158. doi: 10.1002/wnan.<br />

11. Agrawal A, Min D, Singh N, et al. Functional delivery of siRNA in mice<br />

using dendriworms. ACS Nano 2009;3:2495–2504.<br />

12. Patra CR, Bhattacharya R, Patra S, Basu S, Mukherjee P,<br />

Mukhopadhyay D. Inorganic phosphate nanorods are a novel<br />

fl uorescent label in cell biology. J. Nanobiotechnol. 2006;4:11.<br />

13. Wendell D, Jing P, Geng J, et al. Translocation of double stranded<br />

DNA through membrane adapted phi29 motor protein nanopore. Nat.<br />

Nanotechnol. 2009;4(11):765–72.<br />

14. Guo KT, Paul A, Schichor C, Ziemer G, Wendel HP. CELL-SELEX:<br />

Novel perspectives of aptamer-based therapeutics. Int. J. Mol. Sci.<br />

2008;9:668–78.<br />

15. Tsai HA, Ma K, Wang C, et al. Development of integrated protection<br />

for a miniaturized drug delivery system. Smart Mater. Struct. 2007;16,<br />

S295–9.<br />

16. Atanasoska L, Warner RW, Arney MS, Weber J. Drug eluting medical<br />

devices having porous layers. 20090029077, 01/29/2009.<br />

17. Nanoprobes hit targets in tumors, could lessen chemo side effects,<br />

December 15th, 2009; http://www.nanotech-now.com/news.cgi?<br />

story_id=35822<br />

18. Xu ZP, Zeng QH, Lu GQ, Yu AB. Inorganic nanoparticles as carriers for<br />

effi cient cellular delivery. Chem. Eng. Sci 2006; 61: 1027–40.<br />

19. Ambrosi A, Airò F, Merkoçi A, Enhanced gold nanoparticle based ELISA<br />

for breast cancer biomarker, Analytical Chemistry, Anal. Chem. 2010;<br />

82:1151–6.<br />

20 Paradise JK, Diliberto G, Tisdale A, Kokkoli E. Exploring emerging<br />

nanobiotechnology drugs and medical devices. Food Drug Law J.<br />

2008;63:407–20.<br />

21. Holden CA, Yuan Q, Yeudall WA, Lebman DA, Yang H. Surface<br />

engineering of macrophages with nanoparticles to generate a cellnanoparticle<br />

hybrid vehicle for hypoxia-targeted drug delivery. Int. J.<br />

Nanomed. 2010;5:25–36.<br />

22. Yan M, Du J, Gu Z, et al. A novel intracellular protein delivery platform<br />

based on single-protein nanocapsules. Nat. Nanotechnol. 2010;5:48–53.<br />

23. Cole KA, Huggins J, Laquaglia M, et al. RNAi screen of the protein<br />

kinome identifi es checkpoint kinase 1 (CHK1) as a therapeutic target in<br />

neuroblastoma. Proc. Nat. Acad. Sci. 2011;108(8);3336–41.<br />

24. Johnston KP, Maynard JA, Truskett TM, et al. Concentrated dispersions<br />

of equilibrium protein nanoclusters that reversibly dissociate into active<br />

monomers. ACS Nano DOI: 10.1021/nn204166z, Publication Date<br />

(Web): January 19, 2012.<br />

22 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Bhupinder Singh Sekhon et al.: Nanobiotechnology: An overview of drug discovery, delivery and development<br />

25. Cheng Y, Xu Z, Ma M, Xu T. Dendrimers as drug carriers: Applications in<br />

different routes of drug administration. J. Pharml Sci. 2007;97:123–43.<br />

26. Sonke S, Tomalia DA, Dendrimers in biomedical applications refl ections<br />

on the Field, Advanced Drug Delivery Reviews, 2005;57:2106 – 29.<br />

27. Lu J, Shen SS, Li RR, Lin Q, Zheng MJ. Two-dimensional (2D)<br />

crystallization of engineered proteins: A review. Afr. J. Microbiol. Res.<br />

2012;6(1):7–15.<br />

28. Doktorovova S, Souto EB. Nanostructured lipid carrier-based hydrogel<br />

formulations for drug delivery: A comprehensive review. Expert Opin.<br />

Drug Deliv. 2009;6;165–76.<br />

29. Bondì ML, Craparo EF, Giammona G, et al. Nanostructured lipid carrierscontaining<br />

anticancer compounds: Preparation, characterization, and<br />

cytotoxicity studies. Drug Deliv. 2007;14:61–67.<br />

30. Remote-control closed system invented for inserting radioactive atoms,<br />

July 7, 2009; http://www.vtnews.vt.edu/story.php?relyear=2009&itemno<br />

=521<br />

31. Young Wook Chun, Hojean Lim, Thomas J. Webster, Karen M.<br />

Haberstroh. Nanostructured bladder tissue replacements. WIREs<br />

Nanomed Nanobiotechnol 2011;3:134–45;<br />

32. Shrivastava S. Nanofabrication for drug delivery and tissue engineering.<br />

Digest J. Nanomater. Biostruct 2008;3;257–63.<br />

33. Van Bockstaele F, Holz JB, Revets H. The development of nanobodies<br />

for therapeutic applications. Curr Opin Investig Drugs. 2009;10:1212–24.<br />

34. Roodink I, Franssen M, Zuidscherwoude M, et al. Isolation of targeting<br />

nanobodies against co-opted tumor vasculature. Lab. Invest. 2010;90:<br />

61–67.<br />

35. Sikyta B. Nanobiotechnology in pharmacology and medicine. Ceska<br />

Slov Farm. 2001;50(6):263–6.<br />

36. Sharma D. Nanotechnology, 3 Jan 2008. http://www.authorstream.<br />

com/Presentation/Wanderer-37034-sample-seminar-NANO-ROBOT<br />

-BLOOD-STREAM-NANOROBOT-PERFORMING-CELL-SURGERY<br />

-ARTIFICIAL-as-Education-ppt-powerpoint/<br />

37. Stern E, Vacic A, Rajan NK, et al. Label-free biomarker detection from<br />

whole blood. Nat. Nanotechnol., 2010;5(2):138–42.<br />

39. Wang HN, Vo-Dinh T. Multiplex detection of breast cancer biomarkers<br />

using plasmonic molecular sentinel nanoprobes. Nanotechnol. 2009;20:<br />

65–101.<br />

40. Pavon LF, Okamoto O K. Applications of nanobiotechnology in cancer.<br />

Einstein 2007;5:74–77.<br />

41. Jain KK. Application of nanobiotechnology in cancer therapeutics. In:<br />

Pharmaceutical Perspectives of Cancer Therapeutics, Lu Y, Mahato<br />

RI. (eds.) Springer New York. 2009, pp 245–68.<br />

42. Park JH, von Maltzahn G, Xu MJ, et al. Cooperative nanomaterial<br />

system to sensitize, target, and treat tumors. Proc. Natl. Acad. Sci. USA<br />

2010;107(3):981–6.<br />

43. Namiki Y, Namiki T, Yoshida H, et al. A novel magnetic crystal–lipid<br />

nanostructure for magnetically guided in vivo gene delivery. Nat.<br />

Nanotechnol. 2009;4:598–606.<br />

44. Jain KK. Nanomedicine: application of nanobiotechnology in medical<br />

practice. Med. Princ. Pract. 2008;17;89–101.<br />

45. Kemp MM, Linhardt RJ. Heparin-based nanoparticles. Wiley Interdiscipl<br />

inary Reviews: Nanomed. Nanobiotechnol 2010;2:77–87.<br />

46. Kang HG, Tokumasu F, Clarke M. Probing dynamic fl uorescence<br />

properties of single and clustered quantum dots toward quantitative<br />

biomedical imaging of cells. Wiley Interdisciplinary Reviews: Nanomed.<br />

Nanobiotechnol. 2010;2:48–58.<br />

47. Nano World: Nano for artifi cial kidneys, September 8, 2005; http://www.<br />

physorg.com/news6334.html; Nano Medical Products and Devices;<br />

http://www.scribd.com/doc/21052856/Nano-Medical-Products-and-<br />

Devices<br />

48. Lee KH, Kim DJ, Min BG, Lee SH. IFMBE Proceedings: World Congress<br />

on Medical Physics and Biomedical Engineering 2006 , August 27–<br />

September 1, 2006 COEX Seoul, Korea “Imaging the Future Medicine”,<br />

R. Magjarevic and J. H. Nagel, Springer Berlin Heidelberg 2007,<br />

pp 279–82.<br />

49. Veerapandian M, Yun K. The State of the art In biomaterials as<br />

nanobiopharmaceuticals. Digest J. Nanomater. Biostruct. 2009;4;243–62.<br />

50. Kumar A, Gupta OP, Malhotra P. Nanotechnology: Its application in<br />

biopharmacentics. The Pharma Review, November 2009, p74; http://<br />

www.kppub.com<br />

51. Barton RW, Tatake JG, Diwan AR. Nanoviricides - A novel approach<br />

to antiviral therapeutics. In: Bionanotechnology II: Global prospects,<br />

Reisner DE (ed.) CRC Press, 2011.<br />

52. Seĭfulla RD, Timofeev AB, Ordzhonikidze ZG, et al. Nanotechnology<br />

applications in pharmacology. Eksp Klin Farmakol. 2008;71;61–69.<br />

53. Huang B, Tang S, Desai A, et al. Human plasma-mediated hypoxic<br />

activation of indolequinone-based naloxone pro-drugs. Bioorg. Med.<br />

Chem. Lett. 2009;19;5016–20.<br />

54. Jain KK. The role of nanobiotechnology in the development of<br />

personalized medicine. Med. Princ. Pract. 2011; 20: 1–3.<br />

55. Jain KK Role of nanobiotechnology in the development of personalized<br />

medicine. Nanomedicine 2009;4:249–52.<br />

56. Santra S, Dutta D, Walter G, Moudgil BM. Fluorescent nanoparticle<br />

probes for cancer imaging. Technol. Cancer Res. Treat. 2005;4(6),<br />

583–712.<br />

57. Gelain F. Novel opportunities and challenges offered by nanobiomaterials<br />

in tissue engineering. Int. J. Nanomed. 2008;3(4):415–24.<br />

58. Jain KK. Nanobiotechnology in molecular diagnostics: Current<br />

Techniques And Applications, Taylor & Francis, 2006.<br />

59 Baratchi S, Kanwar RK, Khoshmanesh K, et al. Promises of<br />

nanotechnology for drug delivery to brain in neurodegenerative<br />

diseases. Curr. Nanosci. 2009;5(1):15–25.<br />

60. Hearty S, Leonard P, O’Kennedy R. Nanomedicine, Barcodes check out<br />

prostate cancer. Nat. Nanotechnol. 2010;5(1):9–10.<br />

61. Hone J, Kam L. Nanobiotechnology: Looking inside cell walls. Nat.<br />

Nanotechnol. 2007;2:140–1.<br />

62. Zhou H, Chen J, Sutter E, Feygenson M, Aronson MC, Wong SS. Waterdispersible,<br />

multifunctional, magnetic, luminescent silica-encapsulated<br />

composite nanotubes. Small 2010;6(3):412–20.<br />

63. Boisseau P, Houdy P, Lahmani M. (eds.). Nanoscience: Nanobiotechnology<br />

and Nanobiology, Berlin: Springer, 2009.<br />

64. Jokerst JV, McDevitt JT. Programmable nano-bio-chips: Multifunctional<br />

clinical tools for use at the point of care. Nanomed. 2010;5(1):<br />

143–55.<br />

65. Quick blood tests by using a nanodevice. 9 Apr 2009. http://www.<br />

nanomedicinecenter.com/article/quick-blood-tests-by-using-ananodevice/<br />

66. Modi S, Swetha MG, Goswami D, Gupta GD, Mayor S, Krishnan Y. A<br />

DNA nanomachine that maps spatial and temporal pH changes in living<br />

cells. Nat. Nanotechnol 2009;4:325–30.<br />

67. D’Silva I. Recombinant technology and nanoscience. Int. J. Phys. Sci.<br />

2011;6(13):3007–15.<br />

68. Ortiz R, Prados J, Consolación Melguizo C, et al. 5-Fluorouracilloaded<br />

poly(ε-caprolactone) nanoparticles combined with phage E<br />

gene therapy as a new strategy against colon cancer Int. J. Nanomed.<br />

2012;7:95–107.<br />

69. http://www.novosense.se/Research.htm<br />

70. Nanobiotechnologies- Applications, Markets and Companies, Feb. 1,<br />

2012. http://fi nance.yahoo.com/news/Nanobiotechnologies-prnews-<br />

3940639396.html?x=0<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 23


Nasal drug delivery–a review<br />

Twarita Deshpande* 1 , Rajashree Masareddy 2 , Archana Patil 1<br />

1 Rani Chennamma College of Pharmacy, Belgaum<br />

2 College of Pharmacy, K.L.E. University, Belgaum<br />

ABSTRACT<br />

This paper discusses the theory of mucoadhesion along with various approaches to improve nasal absorption by<br />

the use of mucoadhesive polymers and absorption enhancers. An account of various mucoadhesive polymers is<br />

also given. A note on absorption enhancers has been included. Finally the growing market for nasal drug delivery<br />

is discussed.<br />

INTRODUCTION<br />

Nasal drug delivery for systemic effects has<br />

been practiced since ancient times. In modern<br />

pharmaceutics, the nose had been considered<br />

primarily as a route for local drug<br />

delivery. The last two decades heralded a<br />

number of advances in pharmaceutical<br />

biotechnology resulting in possibilities for<br />

large-scale productions of biopharmaceuticals<br />

especially proteins and peptides. The<br />

inability to administer these drugs by routes<br />

other than parenteral injection motivated<br />

scientists to explore other possibilities such<br />

as pulmonary and nasal administration. The<br />

initial enthusiasm was soon confronted<br />

with disappointing in vivo results showing<br />

poor bioavailabilitiy, typically in the order<br />

of 5–10% for large molecules. 1 On the<br />

other hand, very good results were obtained<br />

with small organic molecules, which led<br />

to the successful development of a number<br />

of products currently on the market<br />

list of products that is steadily increasing.<br />

Examination of the causes of failure led<br />

to the conclusion that the short residence<br />

time of the formulation within the nasal<br />

cavity coupled to the low permeability of<br />

the latter did play signifi cant roles. Consequently,<br />

the attention shifted to the evaluation<br />

of mucoadhesive polymers, some<br />

of which would even demonstrate additional<br />

permeation-enhancing capabilities. 2,3<br />

The encouraging results and the desire to<br />

overcome some new challenges stimulated<br />

the development of new generations of<br />

polymers based on pH, thermal responsiveness,<br />

4 modifi ed existing polymers having<br />

improved bioadhesive or permeationenhancing<br />

properties. 5,6,7 Even though a<br />

number of challenges are still to be overcome,<br />

especially with respect to toxicity, the<br />

potential of nasal drug delivery (NDD),<br />

including the ability to target drugs across<br />

the blood–brain barrier (BBB), are very<br />

high and continues to stimulate academic<br />

and industrial research groups so that we<br />

will keep witnessing increasing number of<br />

advanced nasal drug delivery products. To<br />

optimize nasal administration, bioadhesive<br />

hydrogels, Bioadhesive microspheres (dextran,<br />

albumin and degradable starch) and<br />

liposomes have been studied.<br />

ANATOMY<br />

The nasal cavity is divided into two<br />

halves by the nasal septum and extends<br />

posteriorly to the nasopharynx, while the<br />

most anterior part of the nasal cavity, the<br />

nasal vestibule, opens to the face through<br />

the nostril. The atrium is an intermediate<br />

region between the vestibule and the<br />

respiratory region. The respiratory region,<br />

the nasal conchae or turbinates, which<br />

Review Ar cle<br />

Received Date : 08-08-2011<br />

Revised Date : 11-02-2012<br />

Accepted Date : 14-02-2012<br />

DOI: 10.5530/rjps.2012.1.4<br />

Address for<br />

correspondence<br />

Twarita D. Deshpande<br />

Department of Pharmaceutics<br />

Rani Chennamma College of<br />

Pharmacy<br />

Vaibhav Nagar, Belgaum<br />

Pin Code-590010<br />

E-mail: twarita.deshpande@<br />

gmail.com<br />

Mobile: +91 9886181824<br />

www.rjps.in<br />

24 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


occupies the major part of the nasal cavity, possesses<br />

lateral walls dividing it into 3 sections: the superior,<br />

middle and inferior nasal turbinate fi g 1. These folds<br />

provide the nasal cavity with a very high surface area<br />

compared to its small volume.<br />

The epithelial cells in the nasal vestibule are<br />

stratifi ed, squamous and keratinized with sebaceous<br />

glands. Due to its nature, the nasal vestibule is very<br />

resistant to dehydration and can withstand noxious<br />

environmental substances and limits permeation<br />

of substances. The atrium is a transitional epithelial<br />

region with stratifi ed, squamous cells anteriorly<br />

and pseudostratifi ed columnar cells with microvilli<br />

posteriorly. Pseudostratifi ed columnar epithelial cells<br />

interspersed with goblet cells, seromucus ducts, the<br />

openings of sub epithelial seromucus glands cover<br />

the respiratory region (the turbinates). Furthermore,<br />

many of these cells possess actively beating cilia with<br />

microvilli. Each ciliated cell contains about 100 cilia,<br />

while both ciliated and nonciliated cells possess about<br />

300 microvilli each. 8<br />

PHYSIOLOGY<br />

The nasal physiologic functions, such as warming and<br />

humidifi cation, are vital for upper airway function. It has<br />

been estimated that an adult inspires up to 10,000 liters<br />

of air daily.<br />

Figure 1: Anatomy of Nasal Cavity.<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

Nasal breathing is healthy breathing as the air is treated<br />

in many ways by the structures of the nose, paranasal<br />

sinuses and the peculiarities of their lining mucosa.<br />

1. Filtering the air<br />

Filtration of environmental particles occurs fi rst in the<br />

nasal cavity. Nasal mucus traps incoming particulate<br />

matter. The largest particles are fi ltered by nasal hairs<br />

(vibrissae).<br />

2. Moistening the air<br />

Humidifi cation is another important process of nasal<br />

physiology. The nasal cavity is covered with a highly<br />

vascular mucosa that warms and humidifi es incoming<br />

air, increasing the relative humidity to 95% before air<br />

reaches the nasopharynx.<br />

3. Warming the air<br />

Inhaled air must have a temperature between at least<br />

33 and 35 degree Celsius to not cause pathological<br />

reactions at the level of the alveoli. Again, by the<br />

turbulence, the cold air is forced to make contact with<br />

the warm surface of the mucosa and thus heated during<br />

its passage. A number of nasal neurovascular refl exes<br />

occur as well. If needed, underlying capillaries will dilate<br />

and warm up the upper laying mucosa, giving more heat<br />

to the passing air.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 25


4. The sense of smell<br />

Nasal aerodynamics also contributes to the olfactory<br />

system. In addition, the active process of sniffi ng allows<br />

environmental particles to reach the olfactory system<br />

located at the skull base.<br />

Even the smallest particles are detected by the olfactory<br />

receptors, warning us about danger, food, or any other<br />

biologically meaningful sign detectable through the<br />

sense of smell.<br />

5. The nasal cavity as a sound box<br />

The nose and sinuses serve as contributing factors in<br />

voice modifi cation. Authors have noted that nasal<br />

aerodynamics may have a role in modifying highfrequency<br />

sounds and consonants. The resonance<br />

created within the nasal cavity is characteristic, similar<br />

to a fi nger print and for each person is different (except<br />

in identical twins). Nasal pathologies such as polyps or<br />

rhinitis will directly infl uence the resonance spectrum<br />

and we will”hear“that the person has a cold or something<br />

has changed in his/her voice. 9<br />

MUCOADHESION AS A STRATEGY TO<br />

IMPROVE SYSTEMIC DRUG DELIVERY<br />

VIA THE NASAL ROUTE<br />

Several theories have been put forward to explain the<br />

mechanism of polymer-mucus interactions that lead to<br />

mucoadhesion. To start with, the sequential events that<br />

occur during bioadhesion include an intimate contact<br />

between the bioadhesive polymer and the biological<br />

tissue due to proper wetting of the bioadhesive surface<br />

and swelling of the bioadhesive. Following this is the<br />

penetration of the bioadhesive into the tissue crevices,<br />

interpenetration between the mucoadhesive polymer<br />

chains and those of the mucus. Subsequently low<br />

chemical bonds can become operative. 10, 11 Hydration of<br />

the polymer plays a very important role in bioadhesion.<br />

There is a critical degree of hydration required for<br />

optimum bioadhesion. If there is incomplete hydration,<br />

the active adhesion sites are not completely liberated<br />

and available for interaction. An alternative approach to<br />

the use of chemical enhancers to improve nasal drug<br />

absorption is to increase the duration of formulation<br />

residence within the nasal cavity. This is achieved by<br />

the use of bioadhesive polymers. Apart from these<br />

synthetic and natural polymers, there is now a new class<br />

of promising compounds, the lectins, often referred<br />

to as second-generation mucoadhesive materials.<br />

These are non-immunogenic proteins or glycoproteins<br />

capable of specifi c recognition and reversible binding<br />

to carbohydrate moieties of complex glycoconjugates<br />

without altering the covalent nature of any of the<br />

recognized glycosyl ligands. The use of mucoadhesives<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

can solve a number of problems encountered in<br />

controlled drug delivery. It localizes the formulation at<br />

a particular region in the body, thereby improving the<br />

bioavailability of drugs with low bioavailability. The<br />

increased contact time and localization of the drug<br />

due to the strong interaction between the polymer<br />

and mucus is essential for the modifi cation of tissue<br />

permeability. Furthermore, enzymatic activity can be<br />

locally inhibited to improve the bioavailability of drugs<br />

that are subject to enzymatic degradation. This has<br />

been demonstrated for some mucoadhesive polymers<br />

such as Carbopol 934P and polycarbophil that inhibit<br />

the proteolytic enzyme trypsin, which can thus increase<br />

the stability of co-administered Peptides (Leuben<br />

et al. 1994). Some studies have also demonstrated that<br />

mucoadhesive polymers can also directly interact with the<br />

epithelial tight junctions. 12 The mucoadhesive polymers<br />

reported in the literature are summarized in Table 1.<br />

The theories and mechanisms of bioadhesion are given<br />

Table 2. 13<br />

ADVANTAGES OF MUCOADHESIVE DRUG<br />

DELIVERY SYSTEMS<br />

Mucoadhesive dosage forms have three different advantages<br />

when compared to conventional dosage forms<br />

1. These dosage forms are readily localized in the region<br />

applied to improve and enhance the bioavailability<br />

of drugs. Greater bioavailability of piribedit,<br />

testosterone and its esters, vasopressin, dopamine,<br />

insulin was observed from mucoadhesive dosage<br />

forms when compared to conventional dosage<br />

forms.<br />

Table 1: Mucoadhesive Polymers<br />

Polymer Bioadhesive property<br />

Carboxymethyl cellulose<br />

3<br />

Carbopol 934<br />

3<br />

Polycarbophil<br />

3<br />

Tragacanth<br />

3<br />

Poly(acrylic acid/divinyl benzene)<br />

3<br />

Sodium alginate<br />

3<br />

Hydroxy ethyl cellulose<br />

3<br />

Gum Karaya<br />

3<br />

Gelatin<br />

3<br />

Guar gum<br />

2<br />

Thermally modifi ed starch<br />

2<br />

Pectin<br />

1<br />

Polyvinyl Pyrrolidone<br />

1<br />

Acacia<br />

1<br />

Polyethylene glycol<br />

1<br />

Psyllium<br />

1<br />

Amberlite-200 resin<br />

1<br />

Hydroxy propyl cellulose<br />

1<br />

Chitosan<br />

1<br />

Hydroxy ethyl methacrylate<br />

1<br />

NOTE: 3- Excellent, 2- Fair, 1- Poor.<br />

26 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


2. These dosage forms facilitate intimate contact of the<br />

formulation with the underlying absorption surface.<br />

This allows modifi cation of tissue permeability for<br />

absorption surface. This allows modifi cation of tissue<br />

permeability for absorption of macromolecules, such<br />

as peptides and proteins. Inclusion of penetration<br />

enhancers such as sodium glycholate, sodium<br />

taurocholate and L-lysophosphotidyl choline (LPC)<br />

and protease inhibitors in mucoadhesive dosage<br />

forms resulted in better absorption of peptides and<br />

proteins.<br />

3. Mucoadhesive dosage forms also prolong residence<br />

time of the dosage form at the site of application and<br />

absorption to permit once or twice a day dosing. 14<br />

The absorption enhancement mechanisms can be<br />

grouped into two classes:<br />

NASAL ABSORPTION ENHANCERS<br />

Physicochemical effects: Some enhancers can alter the<br />

physicochemical properties of a drug in the formulation.<br />

This can happen by altering the drug solubility, drug<br />

partition coeffi cient, or by weak ionic interactions with<br />

the drug.<br />

Membrane effects<br />

Many enhancers show their effects by affecting the nasal<br />

mucosa surface. 15 Nasal absorption enhancers involve<br />

two main classes. The most important group involve<br />

microspheres, liposome’s and gels that have been utilized<br />

as drug carriers in the past few years. The second group<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

Table 2: Theories and Mechanisms of Bioadhesion<br />

Theory Mechanism of bioadhesion Comments<br />

Electronic theory Attractive electrostatic forces between<br />

Electrons transfer occurs between the two<br />

glycoprotein mucin network and the<br />

forming a double layer of electric charge at<br />

bioadhesive material.<br />

the surface<br />

Wetting theory Ability of bioadhesive polymer to spread<br />

Spreading coeffi cient of polymers must be<br />

and develop intimate contact with the<br />

positive. Contact angle between polymer and<br />

mucous membrane.<br />

cells must be near to zero.<br />

Adsorption theory Surface force resulting in chemical<br />

Strong primary force: covalent bonds. Weak<br />

bonding.<br />

secondary forces: hydrogen bonds and van<br />

der Waal’s forces<br />

Diffusion theory Physical entanglement of mucin strands<br />

For maximum diffusion and best adhesive<br />

and fl exible polymer chains.<br />

strength, solubility parameters of the<br />

bioadhesive polymer and the mucus<br />

glycoproteins must be similar<br />

Mechanical theory Adhesion arises from an interlocking of<br />

Rough surfaces provide an increased surface<br />

liquid adhesive into irregularities on<br />

area available for interaction along with an<br />

the rough surface.<br />

enhanced viscoelastic and plastic dissipation<br />

of energy during joint failure, which are more<br />

important in the adhesion process than a<br />

mechanical effect.<br />

Fracture theory Analyses the maximum tensile stress<br />

Does not require physical entanglement of<br />

developed during attachment of the<br />

bioadhesive polymer chains and mucous<br />

transmucosal DDS from the mucosal<br />

strands, hence it is appropriate to study the<br />

surface<br />

bioadhesion of hard polymers which lack<br />

fl exible chains<br />

will be discussed under the heading other penetration<br />

enhancers.<br />

Dextran microspheres<br />

Illum et al. introduced well-characterized bioadhesive<br />

microspheres for prolonging the residence time in<br />

the nasal cavity. The slowest clearance was detected<br />

for DEAE-dextran, where 60% of the delivered<br />

dose was still present at the deposition site after 3 h. 16<br />

However, these microspheres were not successful in<br />

promoting insulin absorption in rats. The insulin was<br />

too strongly bound to the DEAE groups to be released<br />

by a solution with an ionic strength corresponding to<br />

physiological conditions. Structural changes due to the<br />

lyophilization process were observed in spheres with<br />

insulin incorporated, which probably further decreased<br />

the release rate. 17<br />

Degradable starch microspheres (DSM)<br />

DSM is the most frequently used microsphere system<br />

for nasal drug delivery and has been shown to improve<br />

the absorption of insulin, gentamicin, human growth<br />

hormone, metoclopramide and desmopressin. Insulin<br />

administered in DSM to rats resulted in a rapid dosedependent<br />

decrease in blood glucose. DSM as a<br />

delivery system for insulin (2 IU.kg_1 ) has also been<br />

tested in sheep. The absolute bioavailability was 4.5%<br />

and the time to reach maximum effect, i.e., a 50%<br />

decrease in plasma glucose, was 60 min. 18 Studies in<br />

rabbits have demonstrated that DSM does not induce<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 27


serious hystopathological changes to the nasal mucosa.<br />

Moreover, the DSM was well tolerated by 15 healthy<br />

volunteers and did not cause signifi cant changes in<br />

mucociliary transport. The effect of starch microspheres<br />

on the absorption enhancing effi ciency of various<br />

enhancer systems with insulin after application in the<br />

nasal cavity of the sheep was investigated. The DSM<br />

was shown to synergistically increase the effect of the<br />

absorption enhancers on the transport of the insulin<br />

across the nasal membrane. 19<br />

Liposomes<br />

Various routes have delivered liposomes. Alpar et al.<br />

studied the potential adjuvant effect of liposomes on<br />

tetanus toxoid, when delivered via the nasal, oral and i.m.<br />

routes compared to delivery in simple solution in relation<br />

to the development of a non-parenteral immunization<br />

procedure, which stimulates a strong systemic immunity.<br />

They found that tetanus toxoid entrapped in DSPC<br />

liposomes is stable and is taken up intact in the gut. The<br />

permeability of liposome entrapping insulin through<br />

the nasal mucosa of rabbit has been studied and<br />

compared with the permeability of insulin solution with<br />

or without pre-treatment by sodium glycocholate (GC).<br />

A comparison of the insulin solution and liposome<br />

suspension showed that the liposome had permeated more<br />

effectively after pre-treatment by GC. The relationship<br />

between the rigidity of the liposomal membrane and<br />

the absorption of insulin after nasal administration of<br />

liposomes modifi ed with an enhancer containing insulin<br />

was investigated in rabbits. The nasal administration<br />

to rabbits showed high fl uidity at 37°C, caused a high<br />

serum glucose reduction, and the reduction effect lasted<br />

for 8 h. The loading and leakage characteristics of the<br />

desmopressin-containing liposomes and the effect<br />

of liposomes on the nasal mucosa permeation were<br />

investigated. The increase of permeability antidiuresis<br />

of desmopressin through the nasal mucosa occured in the<br />

order positively charged liposomes > negatively charged<br />

liposomes > solution. The potential of liposomes as an<br />

intranasal dosage formulation or topical application of<br />

5 (6)-carboxyfl uorescein (CF) was investigated in rats. CF<br />

was rapidly absorbed into the systemic circulation and<br />

no adhesion of CF to the nasal mucosa was observed.<br />

Liposomes suppress drug absorption into the systemic<br />

circulation and concurrently increase drug retention in<br />

the nasal cavity. 20<br />

Gels<br />

Chitin and chitosan have been suggested for use as<br />

vehicles for the sustained release of drugs. Indomethacin<br />

and papaverine hydrochloride were used as model drugs<br />

in gel formulations. It was reported that chitin was able<br />

to control the release of the active agents from gel<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

formulation as compared to the powder formulation.<br />

A similar study done later by Lehr et al. showed that<br />

cationic polymer chitosan was fairly mucoadhesive in<br />

comparison to polycarbophil as a reference substance.<br />

They suggested that a strict distinction should be made<br />

between mucoadhesive of dry polymers on a wet tissue<br />

in air and mucoadhesion of a swollen hydro gel in the<br />

presence of a third liquid phase. 21 Nasal absorption of<br />

nifedipine from gel preparations, PEG 400, aqueous<br />

carbopol gel and carbopol-PEG has been studied in<br />

rats. Nasal administration of nifedipine in PEG resulted<br />

in rapid absorption and high Cmax; however, the<br />

elimination of nifedipine from plasma was very rapid.<br />

The plasma concentration of nifedipine after nasal<br />

administration in aqueous carbopol gel formulation was<br />

very low. The use of PEG 400 in high concentration<br />

in humans should be considered carefully because PEG<br />

400 is known to cause nasal irritation in concentrations<br />

higher than 10%. 22<br />

OTHER PENETRATION ENHANCERS<br />

Cyclodextrin<br />

Several compounds have been investigated for their<br />

nasal absorption enhancement potential using cyclodextrins<br />

as the optimisers. The most studied types are:<br />

cyclodextrins cyclodextrin-cyclodextrin methylcyclodextrin<br />

and Hydroxypropyl-cyclodextrin. Only cyclodextrin<br />

is a compendial substance and is being considered<br />

forGRAS (generally recognized as safe) status. Merkus<br />

et al. reported a study, which investigated the effects of<br />

a dimethyl-cyclodextrin (DM-CD) powder formulation<br />

on intranasal insulin absorption in healthy subjects<br />

and patients with insulin-dependent diabetes mellitus<br />

(IDDM). Mean absolute bioavailabilities of 3.1% and<br />

5.1% were achieved in healthy subjects and diabetics,<br />

respectively. 23<br />

Fusidic acid derivatives<br />

Sodium tauro-24, 25-dihydrofusidate (STDHF) is the<br />

most extensively studied among the derivatives of<br />

fusidic acid. On the basis of its characteristics STDHF<br />

was considered a good candidate for the transnasal<br />

delivery of drugs such as insulin, growth hormone and<br />

octreotide. Lee et al. determined the radioimmunoactive<br />

bioavailability of intranasal salmon calcitonin in<br />

10 healthy human volunteers. The improved nasal<br />

absorption of calcitonin in the presence of STDHF<br />

showed a limited transient irritation of the nasal mucosa<br />

in some subjects. Hedin et al. studied the intranasal<br />

administration of human growth hormone (hGH)<br />

in combination with STDHF at 1% concentration<br />

in patients with hGH defi ciency. They found that in<br />

combination with STDHF, the plasma peak of hGH<br />

28 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


was similar to the endogenous peak. Laursen et al. used a<br />

formulation approach in determining the absorption of<br />

growth hormone in human subjects using didecanoyl-<br />

L-phosphotidylcholine (DDPC) as an enhancer with<br />

different concentrations: 0, 4, 8, and 16%. They<br />

concluded that increasing the relative concentration of<br />

DDPC increases the absorption of nasally administered<br />

hGH. Drejer et al. studied intranasal administration of<br />

insulin with DDPC in healthy human volunteers. They<br />

found that intranasal insulin was absorbed in a dose<br />

dependent manner with slight or no nasal irritation. 24<br />

Phosphatidylcholines (PC)<br />

Phosphatidylcholines are surface-active ampiphilic<br />

compounds produced in biological membranes and<br />

liposomes. Several reports have appeared in the<br />

literature showing that these phospholipids can be used<br />

as enhancers for systemic nasal drug delivery. Newman<br />

et al. investigated the distribution of a nasal insulin<br />

formulation containing DDPC labelled with 99m<br />

Tc-human serum albumin (99m Tc-HAS) in human<br />

volunteers. From the scintigraphic data, the entire dose<br />

from the spray was shown to be deposited in the nasal<br />

cavity with no deposition in the lungs.<br />

The Novo Nordisk study group reported encouraging<br />

results following the nasal administration of an insulin/<br />

DDPC microemulsion formulation in human volunteers.<br />

The study demonstrated good absorption of insulin<br />

whilst preventing or minimizing nasal irritation.<br />

Bile salts and surfactants<br />

Commonly used salts are sodium cholate sodium<br />

deoxy cholate, sodium glycocholate (GC), sodium<br />

taurocholate (TC), sodium taurodeoxycholate (TDC),<br />

and sodiumglycodeoxycholate (GDC). Several studies<br />

indicate that bile salts can be good optimisers in nasal<br />

drug products, there are some reports indicating that<br />

bile salts cause nasal irritation when used above a<br />

concentration of 0.3%. Yokosuka and co-workers<br />

reported a study in which healthy volunteers are dosed<br />

nasally with solution formulation containing insulin<br />

and 1%sodiumglycocholate. Signifi cant decreases in<br />

serum glucose concentrations were observed and<br />

there was a positive correlation between the peak<br />

serum insulin levels and the dose of insulin applied.<br />

Hirata et al. investigated the effi cacy of nasal insulin<br />

formulation containing 1% SGC in healthy volunteers<br />

and diabetic patients. The nasal formulation resulted in<br />

rapid increases in serum insulin levels and decreases in<br />

blood glucose levels in healthy volunteers and diabetics.<br />

Moses and colleagues showed that co administration<br />

of 1% sodium deoxycholate (SDC) enhanced the<br />

intranasal absorption of insulin administered to<br />

human volunteers. High inter-subject variability was<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

observed. Frauman compared the effects of intranasal<br />

and subcutaneous insulin on fasting and post-prandial<br />

blood insulin and glucose concentrations in non-obese<br />

patients with non-insulin-dependent diabetes mellitus<br />

(NIDDM). A nasal solution formulation of insulin<br />

and 1% SGC, administered as a spray, resulted in a<br />

monophasic increase in serum insulin levels. Salzman<br />

et al. investigated the effi cacy of 1% laureth-9 in<br />

enhancing the nasal absorption of insulin in patients<br />

with IDDM and non-diabetic controls. Insulin was<br />

shown to be rapidly absorbed via the nasal route<br />

lowering plasma glucose levels to 50% of basal values<br />

after 45 min in normal subjects compared to 50% in<br />

120 min in diabetics. Paquot et al. investigated the<br />

metabolic and hormonal consequences of an intranasal<br />

insulin formulation administration containing 0.25%<br />

laureth-9 in healthy volunteers. Increase in plasma insulin<br />

levels from 5 to 38 mU.I –1 at 15 min with decreases in<br />

blood glucose concentration from 4.4 to 3.2 mmol.I –1<br />

at 45 min.<br />

DRUG ABSORPTION<br />

The fi rst step in the absorption of drugs from the nasal<br />

cavity is passage through the mucus. Small, uncharged<br />

particles easily pass through this layer. However,<br />

larger or charged particles may fi nd it more diffi cult to<br />

cross. Mucin, the principal protein in the mucus, has<br />

the potential to bind to solutes, hindering diffusion.<br />

Additionally, structural changes in the mucus layer<br />

are possible as a result of environmental changes (i.e.,<br />

pH, temperature, etc) 25 Subsequent to a drug’s passage<br />

through the mucus, there are several mechanisms for<br />

absorption through the mucosa. 26 These include<br />

1. Paracellular Transport Mechanism-It mainly uses<br />

an aqueous mode of transport. Usually the drug<br />

passes through the tight junctions and the open<br />

clefts of the epithelial cells present in the nasal<br />

mucosa. It is a relatively passive mode of transport.<br />

Of Compounds, which are highly hydrophilic in<br />

nature and/or low molecular are most appropriate<br />

for paracellular transport.<br />

2. Transcellular Transport Mechanism-It mainly<br />

encompasses transport via a lipodal route. Small<br />

lipophilc compounds or larger molecules are usually<br />

transported by this route. The transport across nasal<br />

mucosa is mainly a function of lipophilc nature of<br />

the drug. 27<br />

FORMULATION ISSUES<br />

Nasal drug absorption is affected by molecular weight,<br />

particle size, formulation pH, pKa of molecule, and<br />

delivery volume among other formulation characteristics.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 29


Molecular weight still presents the best correlation to<br />

absorption. 28 The apparent cut-off point for molecular<br />

weight is approximately 1,000, with molecules less than<br />

1,000 having better absorption. Shape is also important.<br />

Linear molecules have lower absorption than cyclicshaped<br />

molecules. Additionally, particles should be larger<br />

than 10 mm, otherwise the drug may be deposited in the<br />

lungs. 29 Hydrophilicity has also been found to decrease<br />

drug bioavailability. 30<br />

Another formulation factor important for absorption<br />

is pH. Both the pH of the nasal cavity and pKa of<br />

a particular drug need to be considered to optimize<br />

systemic absorption. Nasal irritation is minimized<br />

when products are delivered with a pH range of<br />

4.5 to 6.5. Also, volume and concentration are<br />

important to consider. The delivery volume is limited<br />

by the size of the nasal cavity. An upper limit of<br />

25 mg/dose and a volume of 25 to 150mL/nostril has<br />

been suggested. 31<br />

FDA requirements state all nasal drug products sold<br />

in the United States are to be manufactured as sterile<br />

or preserved products. Depending on the drug and/or<br />

formulation characteristics, sterility may be accomplished<br />

via aseptic fi lling processes, terminal sterilization, or both.<br />

Sterility must also be maintained throughout a product’s<br />

shelf life. Unit-dose delivery systems or specialized<br />

multidose delivery systems may be used to ensure<br />

continued product sterility. Alternatively, preservatives<br />

may be added to prevent bacterial growth. The most<br />

commonly used preservative in nasal formulations has<br />

been benzalkonium chloride. 32 However, adverse events,<br />

such as mucosal swelling, irritation, and ciliostastis, are<br />

associated with its use.<br />

Intranasal vaccine administration has also become<br />

a prevalent area of research. One of the major<br />

formulation challenges for nasal vaccination has been<br />

the development of an appropriate adjuvant to stimulate<br />

desirable immune responses. Some examples of<br />

investigational adjuvants include microparticles, genetic<br />

manipulations of Vibrio cholerae and Escherichia<br />

coli toxins, and CpG oligodeoxynucleotides. The<br />

use of adjuvants is especially important for vaccines<br />

composed of recombinant subunits, synthetic peptides,<br />

and plasmid DNA. For instance, when hepatitis B<br />

surface antigen was given alone to mice, little or no<br />

immunogenicity was observed. However, when given<br />

with CpG or cholera toxin, good immunological<br />

responses were achieved. 33<br />

Drugs have been delivered to the nasal cavity in a variety<br />

of formulations, including powders, topical gels, sprays,<br />

drops, and pledgets. As reviewed elsewhere by Behl et al.,<br />

the dosage form should refl ect the intended therapeutic<br />

use, offer easy administration, and provide chemical<br />

stability for the drug. 34<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

With any formulation for intranasal delivery,<br />

interference with normal physiologic processes should<br />

be avoided. The nose is an important defense system<br />

for environmental hazards. Disruption of its normal<br />

physiology or clearance processes may leave a patient<br />

vulnerable to a variety of hosts. Also, formulations<br />

should be designed with patient comfort and acceptance<br />

in mind. Some drugs and excipients have been found<br />

to cause temporary discomfort upon instillation. This<br />

could potentially decrease patient compliance, especially<br />

when chronic administration is indicated. 35–38<br />

NASAL DELIVERY OF VACCINES<br />

Almost all viral, bacterial and parasitic agents causing<br />

common infectious diseases of the intestinal, respiratory<br />

and genital tract enter and infect through the large<br />

surface area made available by mucosal membranes.<br />

The nasal mucosa is an important arm of the mucosal<br />

immune system since it is often the fi rst point of contact<br />

for inhaled antigens and as a consequence, intranasal<br />

immunisation has emerged as possibly the most effective<br />

route for vaccination for both peripheral and mucosal<br />

immunity.<br />

Recently, Partidos has considered the challenges for<br />

intranasal vaccines. He has listed several reasons why the<br />

nose is an attractive route for immunisation.<br />

• Easily accessible.<br />

• Highly vascularized.<br />

• Presence of numerous microvilli covering the nasal<br />

epithelium generates a large absorption<br />

• Surface.<br />

• After intranasal immunisation, both mucosal and<br />

systemic immune responses can be induced.<br />

• Immune response can be induced at distant mucosal<br />

sites owing to the dissemination of effector immune<br />

cells in the common mucosal immune system.<br />

• The nose can be used for the easy immunization of<br />

large population groups.<br />

• Nasal immunisation does not require needles and<br />

syringes, which are potential sources of infection. 39<br />

The utility of intranasal administration for a variety of<br />

clinical applications to include largely the prophylaxis<br />

of disease. Other uses of nasal vaccination have been<br />

reported. For example, Weiner et al. have suggested a<br />

novel mucosal immunlogical approach to Alzheimer’s<br />

disease. Nasal administration of amyloid-beta peptide<br />

decreased cerebral amyloid burden in a mouse model of<br />

the disease. Recently, live attenuated; cold adapted viral<br />

vaccines have been developed as alternatives to inactivated<br />

vaccines. For example, by growing the infl uenza virus<br />

at 25ºC for long periods or using genetic reassortment<br />

methods, it is possible to produce an organism that will<br />

30 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


eplicate effi ciently at 25–28ºC (the temperature of the<br />

nasal passage) but not at 37ºC (the temperature of the<br />

lungs). Clinical studies on live attenuated virus vaccines<br />

have been well reviewed by Wiselka and Keitel and<br />

Piedra. One major goal of intranasal vaccination with<br />

a replicating virus is to induce secretory and systemic<br />

immune responses that more closely resemble those<br />

provided by the natural infection.<br />

BRAIN TARGETING THROUGH NASAL ROUTE<br />

For some time the Blood Brain Barrier has impeded the<br />

development of many potentially interesting CNS drug<br />

candidates due to their poor distribution into the CNS.<br />

The Blood Brain Barrier is a system of layers of cells at<br />

the cerebral capillary endothelium, the choroids plexus<br />

epithelium, and the arachnoid memberanes, which<br />

are connected by tight junctions and which together<br />

separate the brain and the cerebrospinal fl uid from<br />

blood. 40 Owing to the uniqueness of the nose and<br />

the CNS, the intranasal route can deliver therapeutic<br />

agents to the brain bypassing the Blood brain Barrier. 41<br />

Absorption of drug across the olfactory region of the<br />

nose connection provides a unique feature and superior<br />

option to target drugs to brain. When administered<br />

nasally to the rat, some drugs resulted in CSF and<br />

olfactory bulb drug levels considerably higher than<br />

those following intravenous administration. 42 Evidence<br />

of nose to brain transport has been reported by many<br />

scientists. Many previously abandoned potent CNS<br />

drug candidates promise to become successful CNS<br />

therapeutic drugs via intranasal delivery. Recently,<br />

several nasal formulations, such as ergotamine<br />

(Novartis), sumatriptan (GlaxoSmith-Kline), and<br />

zolmitriptan (AstraZeneca) have been marketed to treat<br />

migraine. Scientists have also focused their research<br />

toward intranasal administration for drug delivery to<br />

the brain, especially for the treatment of diseases, such<br />

as, epilepsy, migraine, emesis, depression and erectile<br />

dysfunction. 43,44 Intranasal delivery does not require<br />

any modifi cation of the therapeutic agents and does<br />

not require that drugs be coupled with any carrier.<br />

A wide variety of therapeutic agents, including both<br />

small molecules and macromolecules can be successfully<br />

delivered to the CNS via intranasal method. 45<br />

ADMINISTRATION DEVICE<br />

Drug therapy requires that administration of the dosage<br />

form be accurate and very reproducible, which therefore<br />

places stringent, demands on the device for nasal drug<br />

delivery. The major mechanism of nasal deposition of<br />

particles is by inertial impaction that occurs following a<br />

change in the direction of airfl ow. Other contributory<br />

mechanisms are gravitational sedimentation and<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

Brownian diffusion. Particle deposition by interception<br />

and electrostatic precipitation are of no importance in<br />

nasal deposition (Kublik & Vidgren 1998). Depending<br />

on the type of formulation, a variety of devices have<br />

been used to deliver drugs intranasally. Devices for liquid<br />

formulations include instillation catheter, droppers,<br />

unit-dose containers, squeezed bottle, pump sprays,<br />

airless and preservative-free sprays, compressed air<br />

nebulizers and metered-dose inhalers (MDIs). Devices<br />

for powder dosage forms include insuffl ators, singledose<br />

and multi-dose powder inhalers and pressurized<br />

MDI. Delivery devices are also available for nasal gels<br />

(Kublik & Vidgren 1998). Squeeze bottle delivery<br />

is another option for nasal drug delivery. However<br />

this technique is not able to deliver a measured dose<br />

of drug.<br />

Their angle of insertion into the nostril can infl uence<br />

the part of the nasal cavity that the formulation<br />

comes in contact with initially and as such the overall<br />

deposition pattern. Metered-dose nebulizers and<br />

metered-dose aerosols are superior to other devices in<br />

terms of accuracy and reproducibility (Dondeti 1996).<br />

1985). The particle size of aerosols is very important<br />

with regard to deposition. Particles greater than 10 ml<br />

are deposited within the upper respiratory tract, those<br />

less than 5 ml are inhaled, and those less than 0.5 ml<br />

are exhaled (Sciarra & Cutie 1990; Sanders et al 1997).<br />

A brief description of various intranasal devices is given<br />

as follows Figure 2–8.<br />

1. Caprujet of midazolam–This is a prepackaged<br />

midazolam in 5mg/ml dosing. It is indicated for<br />

treatment of persistent seizure activity. In situations<br />

where the entire volume might not be appropriate,<br />

the syringe can have a indelible mark made by the<br />

clinician or pharmacist at appropriate dosage so the<br />

parent knows how much to give.<br />

2. Direct-Haler powdered Drug Nasal Delivery–This<br />

system enables nasal delivery of fi ne particles for<br />

improved absorption and targeted delivery. Prevents<br />

risk of pulmonary deposition. Prevents risk of<br />

immediate dose swallowing, reduces taste impact<br />

from nasal use.<br />

3. Mucosal Atomization Device(MAD)–Delivers<br />

intranasal medication in a fi ne mist which enhances<br />

absorption and improves bioavailability for fast and<br />

effective drug delivery.<br />

4. Via Nose Electronic Atomizer–This atomizer<br />

creates tiny, Powerful vortices of aerosolized particles,<br />

thus delivering medication more effectively to the<br />

user. This device has a small LCD screen which is<br />

used to specify dose.<br />

5. BDA Accuspray Nasal Spray System–It is a<br />

single use, prefi llable vaccine delivery system which<br />

dispenses a precise dose of fl u vaccine intranasally in<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 31


Figure 2: Nasal Delivery Dosage Forms and Devices.<br />

Figure 3: Caprujet of Midazolam.<br />

the form of a fi ne mist. Recommended for healthy<br />

individuals in the age group of 5–49.<br />

6. Optinose–This device is based on the concept of<br />

a bi-directional nasal drug delivery device. Recently<br />

optinose received positive results from their phase<br />

II clinical studies for chronic rhinosinusitis and<br />

migraine therapies.<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

Regardless of the method chosen to deliver a nasal<br />

medication, several suggestions are worth mentioning:<br />

• Use a highly concentrated form of the medication to<br />

reduce volume and therefore reduce runoff. 1/4 to<br />

1/3 ml per nostril would be preferred. 1/2 to 1 ml per<br />

nostril is tolerable but there will be some loss as the<br />

volume increases. More than one ml per nostril per<br />

dose should likely be split and delivered over several<br />

cycles separated by 10–15 minutes.<br />

• Always use a method that allows the delivery of a<br />

measured dose (syringe or unit dose pump).<br />

• Use a method that fragments the medication into<br />

fi ne particles so the maximal nasal mucosal surface<br />

is covered and minimal volume runs out the nose or<br />

into the throat (atomizer for liquid, powder needs to<br />

be well distributed).<br />

• Utilize both nostrils to double the surface area for<br />

absorption and halve the volume delivered per<br />

nostril.<br />

• Be knowledgeable of the “dead space” within the<br />

delivery device and account for this dead space when<br />

calculating the volume you will deliver to the patient.<br />

Considerations Regarding Device Selection<br />

When selecting a device for nasal administration, it<br />

must be considered, that the administration volume<br />

is comparable low. For liquids, a volume of 100μl is<br />

32 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Figure 4: Direct-Haler Powdered Drug Nasal Delivery.<br />

Figure 5: Mucosal Atomization Device (MAD).<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 33


Figure 6: Via Nase Electronic Atomizer.<br />

Figure 7: BDA Accuspray Nasal Spray System<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

34 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Figure 8: OptiNose.<br />

optimum per nostrils in a adults, but should be reduced<br />

for children to avoid nasal dripping. Single dose devices<br />

will give best protection for vaccines, but require<br />

highly sophisticated fi lling technology. Multi-dose<br />

spray pumps are option for liquid vaccines, if an in use<br />

microbial contamination of the bottle content can be<br />

prevented. 46<br />

ADVANTAGES OF NASAL DELIVERY<br />

Intranasal delivery is a needle-free, patient-friendly<br />

administration route. Because needles are not involved,<br />

this method of drug delivery is virtually painless. For<br />

patients who fear injections, intranasal administration<br />

offers a more acceptable alternative. Additionally,<br />

the simplicity of nasal delivery would allow for selfadministration<br />

in a home setting. In general, for patients,<br />

the intranasal dosage form provides comfortable, non<br />

threatening, less invasive therapy. This may be especially<br />

important in younger patient populations.<br />

Another major benefi t of intranasal administration, in<br />

contrast to injectables, is that it does not contribute to<br />

biohazardous waste. When the drug has been delivered<br />

intranasally, the administration device may be disposed<br />

of in the normal garbage. There is no need for special<br />

waste containers. Again, this delivery method does not<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

require needles. Hence risk of accidental sticks is not a<br />

concern.<br />

From a pharmacokinetic standpoint, absorption is<br />

rapid, which should provide a faster onset of action<br />

compared to oral and intramuscular administration.<br />

Hepatic fi rst-pass metabolism is also avoided, allowing<br />

increased, reliable bioavailability. In this regard, good<br />

drug candidates for intranasal delivery are those that<br />

undergo extensive fi rst-pass metabolism, display erratic<br />

absorption, or require quick therapeutic onset.<br />

Lastly, patent life of a particular product may be<br />

extended via development of an alternative dosage form,<br />

providing companies the opportunity to maintain their<br />

market share. So from a drug development perspective,<br />

intranasal delivery should stimulate favorable profi t<br />

outcomes. 47<br />

GROWING MARKET FOR NASAL DRUG DELIVERY<br />

Pharmaceutical companies have looked increasingly<br />

towards drug delivery companies for help in lifecycle<br />

management of drugs on the market and with promising<br />

yet hard-to-deliver drugs. The drug delivery market<br />

is currently valued at US$50 billion (or 12.5% of the<br />

global pharmaceutical market) and is expected to reach<br />

US$100 billion by 2005.1 Nasal delivery commands the<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 35


fourth position in market share, with about US$3 billion<br />

in sales, following oral controlled release, pulmonary and<br />

parenteral delivery routes. However, the potential for<br />

growth in this sector is extensive, pending the successful<br />

delivery of proteins and peptides as an alternative to<br />

parenteral delivery. Currently, many nasal drug products<br />

on the market are indicated for the treatment of<br />

local disease such as allergic rhinitis. However, this is<br />

likely to change soon. There are a number of nasally<br />

delivered, systemically acting drugs on the market<br />

in different therapeutic categories, with a growing<br />

number of products in the pipeline. There are many<br />

reasons for this change, including improved patient<br />

compliance (elimination of needles), avoidance of fi rstpass<br />

metabolism and rapid onset of action. Migraine is<br />

a key area where a nasal system (Imitrex ® nasal spray,<br />

GlaxoSmithKline) has provided rapid relief, avoidance<br />

of taking an oral formulation while nauseated, and<br />

pain-free administration circumventing the need for an<br />

injection. Other therapeutic areas where nasal delivery<br />

could provide an alternative to current dosage forms<br />

are crisis situations (seizure and heart attack), erectile<br />

dysfunction, pain management, motion sickness and<br />

psychotropic drugs. Although the market share for nasal<br />

delivery may never take the number one spot enjoyed by<br />

oral controlled release, it remains a drug delivery route<br />

with an enormous potential for growth.<br />

REFERENCES<br />

1. Behl CR, Pimplaskar HK and Sileno AP. Effects of physicochemical<br />

properties and other factors on systemic nasal drug delivery. Adv Drug<br />

Deliv Rev 1998;29:89–116.<br />

2. Mathiowitz E, Chickering DE, Lehr CM. Bioadhesive drug delivery<br />

systems: Fundamentals, Novel approaches and development. First<br />

edition. New York: Marcel Dekker Inc;1999.<br />

3. Dodane V, Khan MA, Merwin JR. Effect of Chitosan on epithelial<br />

permeability and structure. Int J Pharm 1999;182:21–32.<br />

4. Nakamura K, Maitani Y, Lowman AM. Uptake and release of budesonide<br />

from mucoadhesive, pH-sensitive copolymers and their application to<br />

nasal delivery. J Control Release 1999;61:329–5.<br />

5. Schnurch B, Kast CE , Richter MF. Improvement in the mucoadhesive<br />

properties of alginate by the covalent attachment of cysteine. J Control<br />

Release 2001;71:277–5.<br />

6. Wang J, Sakai S, Y D Bi Deguchi, Tabata Y, Morimoto K. Aminated<br />

gelatin as a nasal absorption enhancer for peptide drugs: evaluation<br />

of absorption enhancing effect and nasal mucosa perturbation in rats.<br />

J Pharm Pharmacol 2002;54:181–8.<br />

7. Arora P, Sharma S, Garg S. Permeability issues in nasal drug delivery.<br />

Drug Discov Today 2002;7:967–5.<br />

8. Jime´nez-Castellanos MR, Zia H, Rhodes 19:143–194. CR. Mucoadhesive<br />

drug delivery systems. Drug Dev Ind Pharm 1993;<br />

9. IAH AC Rhinopathologies [online].2007 Available from: URL: www.iah<br />

-online.com/cms/docs/doc26624.pdf<br />

10. Ducheˆne D, Touchard F, Peppas NA. Pharmaceutical and medical<br />

aspects of bioadhesive systems for drug administration. Drug Dev Ind<br />

Pharm 1998;14:283–318.<br />

11. Ch’ng HS, Park H, Kelly P, Robinson JR. Bioadhesive polymers as<br />

platforms for oral controlled drug delivery: II. Synthesis and evaluation<br />

of some swelling, water-insoluble bioadhesive polymers. J Pharm Sci<br />

1985;74:399–405.<br />

12. Michael UI, Norbert V, Renaat K. The biopharmaceutical aspects of<br />

nasal mucoadhesive drug delivery. J Pharm Pharmacol 2001;53:3–22.<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

13. Smart JD. The basics and underlying mechanisms of mucoadhesion.<br />

Adv Drug Delivery 2005;57:1556–8.<br />

14. Mathiowitz E, Chickering DE, Lehr CM. Bioadhesive drug delivery<br />

systems: Fundamentals, Novel approaches and development. First<br />

edition. New York: Marcel Dekker Inc; 1999.<br />

15. Behl CR, Pimplaskar HK, Sileno AP etal. Optimization of systemic<br />

nasal drug delivery with pharmaceutical excipients. Adv Drug Del Rev<br />

1998;29:117–33.<br />

16. llum L, Jorgensen H, Bisgaard H, Krogsgaard O, Rossing N. Bioadhesive<br />

microspheres as a potential nasal drug delivery system. Int J Pharm<br />

1987;39:189–99.<br />

17. Morath LP. Microspheres as nasal drug delivery systems. Adv Drug Del<br />

Rev 1998;29:185–94.<br />

18. Hinchcliffe M, Illum L. Intranasal insulin delivery and therapy. Adv Drug<br />

Del Rev 1999;35:199–234.<br />

19. Vyas SP, Bhatnagar S, Gogoi PJ, Jain NK. Preparation and characterization<br />

of HAS-propranolol microspheres for nasal administration. Int J<br />

Pharm 1991;69:512.<br />

20. Alpar HO, Bowen JC, Brown MRW. Effectiveness of liposomes<br />

adjutants of orally and nasally administered tetanus toxoid. Int J Pharm<br />

1992;88:335–44.<br />

21. Morimoto K, Tabata H, Morisaka K. Nasal absorption of nifedipine from<br />

gel preparations in rats. Chem Pharm Bull 1987;35:3041–4.<br />

22. Merkus FWHM, Verhoef. J.C, Romeijn SG, Schipper NGM. Absorption<br />

enhancing effect of cyclodextrins on intranasally administered insulin in<br />

rats. Pharm Res 1991;8:588–92.<br />

23. Lee WA, Ennis RD, Longenecker JP, Bengtsson P. The bioavailability<br />

of intranasal salmon calcitonin in healthy volunteers with and without a<br />

permeation enhancer. Pharm Res 1994;11:747–50.<br />

24. Selcan T, Erten O, Yekta O. Nasal route and drug delivery systems.<br />

Pharma world sci 2004;26:137–42.<br />

25. Khanvilkar K, Donovan MD, Flanagan DR. Drug transfer through<br />

mucus. Adv Drug Deliv Rev 2001;28:173–93.<br />

26. Marttin E, Verhoef JC, Cullander C, Romeijn SG, Nagelkerke JF,<br />

Merkus FWHM. Confocal laser scanning microscopic visualization of<br />

the transport of dextrans after nasal administration to rats: effects of<br />

absorption enhancers. Pharm Res 1997;14:631–7.<br />

27. Vyas Tk, Shahiwala A, Marathe S, Misra A. Intranasal Drug Delivery for<br />

Brain Targeting. Current Drug Delivery2005;2:1–11.<br />

28. Fisher AN, Brown K, Davis SS, Parr GD, Smith DA. The effect of<br />

molecular size on the nasal absorption of water-soluble compounds in<br />

the albino rat. J Pharm Pharmacol 1987;39:357–62.<br />

29. Jones NS, Quraishi S, Mason JDT. The nasal delivery of systemic<br />

drugs. Int J Clin Pract 1997;51:308–11.<br />

30. Corbo D, Liu J, Chien YW. Drug absorption through mucosal membranes:<br />

effect of mucosal route and penetrant hydrophilicity. Pharm Res<br />

1989;6:848–52.<br />

31. Behl CR, Pimplaskar HK, Sileno AP, deMeireles J, Romeo VD. Effects<br />

of physiochemical properties and other factors on systemic nasal drug<br />

delivery. Adv Drug Delivery Rev 1998;29:89–116.<br />

32. Calcitonin-salmon Nasal spray product information[online]. 2009[cited<br />

2009 Feb]. Available from: URL:http://www.drug.com/pro/calcitoninsalmon-nasal-spray-html<br />

33. Morimoto K, Katsumata H, Yabuta T et al. Evaluation of gelatin<br />

microspheres for nasal and intramuscular administrations of salmon<br />

calcitonin. Eur J Pharm Sci. 2001;13:179–85.<br />

34. Mathiowitz E, Chickering DE, Lehr CM. Bioadhesive drug delivery<br />

systems: Fundamentals, Novel approaches and development. First<br />

edition. New York: Marcel Dekker Inc;1999.<br />

35. Davis SS. Nasal vaccines. Adv Drug Deliv Rev 2001;51:21–42.<br />

36. McCluskie MJ, Davis HJ. Cutting edge: CpG DNA is a potent enhancer<br />

of systemic and mucosal immune responses against hepatitis B<br />

surface antigen with intranasal administration to mice. J Immunol<br />

1998;161:4463–6.<br />

37. Illum L, Davis SS. Intranasal insulin. Clin Pharmacokinet 1992;12:<br />

30–41.<br />

38. van de Donk HJM, Merkus FWHM. Decreases in ciliary beat<br />

frequency due to intranasal administration of propanolol. J Pharm Sci<br />

1982;71:595–6.<br />

39. Mestecky J, Moldoveanu Z, Michalek SM. Current options for vaccine<br />

delivery systems by mucosal routes. J Control Release1997;48:<br />

243–57.<br />

40. Bommer R. Latest advances in nasal drug-delivery technology. Med<br />

Device Technol 1999;10:22–8.<br />

36 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


41. Frey WH II. Intranasal delivery: Bypassing the blood- brain to deliver<br />

therapeutic agents to the brain and spinal cord. Drug delivery technology<br />

2002;5:46–9.<br />

42. Hussain AA. Intranasal drug delivery. Adv Drug Del Rev 1998;29:39–9.<br />

43. Chow HS, Chen Z, Matsuura GT. Direct transport of cocaine from nasal<br />

cavity to the brain following intranasal cocaine administration in rats.<br />

J Pharm Sci 1999;88:754–8.<br />

44. Sakane T, Akizuki M, Yoshida M. Transport of cephalexin to the<br />

cerebrospinal fl uid directly from nasal cavity. J Pharm Pharmacol<br />

1991;43:449–51.<br />

Twarita Deshpande et al.: Nasal Drug Delivery-A Review<br />

45. Talegaonkar S, Mishra PR. Intranasal Delivery: An approach to bypass<br />

the blood brain barrier. Indian J Pharmacol 2004;36:140–7.<br />

46. Leitz m, Marx D, Birkhoff M. Advantages of Intranasal Vaccination<br />

and Considerations on Device Selection. Indian J Pharm Sci 2009;71:<br />

729–31.<br />

47. Wermeling DP, Miller J, Rudy AC. Systematic Intranasal Drug Delivery:<br />

Concepts and Applications. Drug Development & Delivery 2002;2.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 37


Design and optimization of levofl oxacin<br />

gastroretentive tablets<br />

D. Nagendrakumar 2 , S.B. Shirsand* 1 , M.S. Para 2 , A.D. Chauhan 2<br />

1 Department of Pharmaceutical Technology, H.K.E. Society’s M T R Institute of Pharmaceutical Sciences,<br />

Sedam Road, Gulbarga-585105, India<br />

2Department of Pharmaceutics, S.V.E. Trust’s College of Pharmacy, Kallur Road, Humnabad-585330, India<br />

ABSTRACT<br />

In the present study, gastroretentive fl oating tablets of levofl oxacin hemihydrate were designed with objective<br />

of retention of tablet in acidic pH to improve bioavailability with reduction in dosing frequency. Hydroxypropyl<br />

methyl cellulose of different viscosity grades (K4M and K100LV) was used as polymer and sodium bicarbonate<br />

as gas generating agent to reduce fl oating lag time. Tablets were prepared by direct compression method. The<br />

prepared formulations were evaluated for hardness, friability, weight variation, drug content, swelling index,<br />

in-vitro drug release, short-term stability, fl oating lag time and in-vitro buoyancy. A 32 factorial design was applied<br />

to systematically optimize the drug release profi le.<br />

The proportions of release retardant material HPMC K100LV(X ), sodium bicarbonate (X ) were selected as<br />

1 2<br />

independent variables and t (Y ), and t (Y ) were selected as dependent variables. The promising formulation<br />

50% 1 70% 2<br />

containing levofl oxacin hemihydrate (100 mg), HPMC K100LV (100 mg) and sodium bicarbonate (80 mg) has t50% (5.95 h), t (8.52 h) fl oating lag time was only 10.33 sec and released more than 90% drug in 12 h. This study<br />

70% ,<br />

proved that gastroretentive drug delivery system of levofl oxacin hemihydrate was designed using HPMC K100LV,<br />

which provided excellent gastroretentive property, thus improved the bioavailability of drug.<br />

Key words: Levofl oxacin hemihydrate, Gastroretentive fl oating drug delivery systems, Hydroxypropyl methyl<br />

cellulose, 3² factorial design<br />

INTRODUCTION<br />

The real challenge in the development of an<br />

oral controlled-release drug delivery system<br />

is not just to sustain the drug release, but<br />

also to prolong the presence of the dosage<br />

form within the gastrointestinal tract (GIT)<br />

until the drug is completely released at the<br />

desired period of time. 1 Indeed, gastric<br />

drug retention has received signifi cant<br />

interest in the past few decades. Most of<br />

the conventional oral delivery systems have<br />

shown some limitations related to fast<br />

gastric emptying time. 2<br />

Garg and Gupta3 classifi ed the gastroretentive<br />

dosage forms into four main classes: (i)<br />

fl oating systems4 (ii) expandable systems5 (iii) bioadhesive systems6 and (iv) high<br />

density systems. 7 Floating systems are of<br />

two types: effervescent systems, depending<br />

on the generation of carbon dioxide gas<br />

upon contact with gastric fl uids, and noneffervescent<br />

systems. The non-effervescent<br />

systems can be further divided into four<br />

sub-types, including hydrodynamically balanced<br />

systems, 8 microporous compartment<br />

systems, 9 alginate beads 10 and hollow microspheres/microballons.<br />

11 In addition, superporous<br />

hydrogels 12 and magnetic systems 13<br />

were also described.<br />

As suggested by Singh and Kim, 14 fl oating<br />

drug delivery is of particular interest for the<br />

drugs which: (a) act locally in the stomach;<br />

(b) are primarily absorbed in the stomach;<br />

(c) are poorly soluble at an alkaline pH;<br />

Research Ar cle<br />

Received Date : 11-07-2011<br />

Revised Date : 11-02-2012<br />

Accepted Date : 15-02-2012<br />

DOI: 10.5530/rjps.2012.1.5<br />

Address for<br />

correspondence<br />

S B Shirsand<br />

Department of Pharmaceutical<br />

Technology<br />

H.K.E. Society’s M T R Institute<br />

of Pharmaceutical Sciences<br />

Sedam Road<br />

Gulbarga-585105, India<br />

E-mail: shirsand@rediffmail.<br />

com<br />

www.rjps.in<br />

38 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


D.Nagendrakumar et al.: Design and Optimization of Levofl oxacin Gastroretentive Tablets<br />

(d) have a narrow window of absorption and (e) are<br />

unstable in the intestinal or colonic environment.<br />

Levofl oxacin hemihydrate is a broad spectrum<br />

fl uoroquinolone antibiotic. It is approved for use in<br />

the treatment of Helicobacter pylori infection. H. pylori<br />

was recognized as a major gastric pathogen responsible<br />

for chronic active gastritis, duodenal ulcers and gastric<br />

adeno-carcinoma. 15 The bioavailability of levofl oxacin<br />

hemihydrate is above 99% with a plasma half-life of<br />

6–8 h. It is freely soluble in pH 0.6 to 5.8 range. Above<br />

pH 5.8, the solubility increases rapidly to its maximum<br />

at pH 6.7 and above which the solubility decreases and<br />

reaches a minimum value at a pH of approximately<br />

6.9. 16 Thus solubility of the drug is reduced in intestinal<br />

alkaline pH. Hence, it was selected in the present<br />

investigation as a suitable candidate for the design of<br />

gastric fl oating drug delivery system (GFDDS) with an<br />

improved retention time and bioavailability.<br />

In a full factorial design, two factors were studied at all the<br />

possible combinations, by considering it is most effi cient<br />

in estimating the infl uence of individual variables and<br />

their interactions, using minimum experimentation. In the<br />

present investigation, amount of HPMC K100LV (X 1 )<br />

and amount of sodium bicarbonate (X 2 ) were selected<br />

as independent variables in the 3 2 factorial design. The<br />

time required for drug release of 50% (t 50%, Y 1 ) and 70%<br />

(t 70%, Y 2 ) were selected as dependent variables. A statistical<br />

model incorporating interactive and polynomial<br />

terms was utilized to evaluate the response.<br />

MATERIALS AND METHODS<br />

Levofl oxacin hemihydrate was gifted by Zhejiang Starry<br />

Pharmaceutical Ltd., Mumbai (India), hydroxypropyl<br />

methylcellulose K100LV was gifted by Colorcon<br />

Asia Pvt. Limited, Goa (India). Sodium bicarbonate,<br />

magnesium stearate, talc were purchased from (SD fi ne<br />

chem., Mumbai). All the materials were of analytical or<br />

pharmacopoeial grade and used as received.<br />

PREPARATION OF LEVOFLOXACIN HEMIHYDRATE<br />

FLOATING TABLETS<br />

Preparation<br />

Direct compression method has been employed<br />

to prepare gastroretentive tablets of levofl oxacin<br />

hemihydrate using HPMC K100LV as polymer and<br />

sodium bicarbonate as gas generating agent.<br />

Procedure<br />

All the ingredients including drug, polymer and<br />

excipients were weighed accurately according to the each<br />

tablet formulation (Table 1). The polymer and sodium<br />

bicarbonate were thoroughly mixed on a butter paper<br />

with the help of a stainless steel spatula. The mixture was<br />

mixed with drug and then excipients were added in the<br />

order of ascending weights, the mixture was blended for<br />

10 min in an infl ated polyethylene pouch. The prepared<br />

blend of each formulation was compressed on 10-station<br />

rotary tablet punching machine (Clit, Ahmedabad) to<br />

obtain fl at-faced tablet of 8 mm diameter.<br />

EVALUATION OF GASTRORETENTIVE<br />

FLOATING TABLETS<br />

The prepared formulations were evaluated for hardness,<br />

friability, weight variation, drug content, swelling index,<br />

in-vitro drug release, short-term stability, fl oating lag time<br />

and in- vitro buoyancy.<br />

Hardness test<br />

The crushing strength (kg/cm²) of tablets was<br />

determined by using digital hardness tester 17 (Electro<br />

Lab E01). The mean of three determinations were taken.<br />

Friability test<br />

It was determined by weighing 10 tablets (W ) after<br />

Original<br />

dusting, placing them in the friabilator (Electro lab EF-2)<br />

and rotating the plastic cylinder vertically at 25 rpm for<br />

4 min. 18 After dusting, the total remaining weight of the<br />

tablets (W ) was recorded and the percent friability<br />

Final<br />

was calculated according to Equation 1.<br />

Weight Final − WeightOriginal<br />

Percent friability =<br />

× 100 (1)<br />

Weight<br />

Uniformity of weight<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 39<br />

Original<br />

The weight (mg) of each of 20 individual tablets was<br />

determined using an electronic balance (Shimadzu<br />

BL-220H). The weight data from the tablets were<br />

analyzed for sample mean and percent deviation. 18<br />

Uniformity of drug content<br />

Two tablets were powdered in a glass mortar and the<br />

powder equivalent to 1 mg of drug was placed in a<br />

500 ml conical fl ask. The drug was extracted with<br />

500 ml of pH 1.2 solution with vigorous shaking<br />

on a mechanical gyratory shaker (100 rpm) for 1 h.<br />

Absorbance was measured at 293 nm in Shimadzu<br />

UV-1800 spectrophotometer against blank.<br />

In-vitro fl oating study<br />

Floating time was determined by using USP XXIII tablet<br />

dissolution apparatus (Electro lab TDT-08L) at 50 rpm<br />

using 900 ml of 0.1N HCl at 37 ± 0.5°C temperature.<br />

The fl oating duration was the time during which the<br />

tablet remains buoyant and fl oating lag time was the<br />

time between tablet introduction and its buoyancy.


D.Nagendrakumar et al.: Design and Optimization of Levofl oxacin Gastroretentive Tablets<br />

Table 1: Composition of Factorial Design and Check point Formulations of Levofl oxacin Hemihydrate<br />

Ingredients*<br />

Formulation code<br />

(mg/tablet)<br />

F1 F2 F3 F4 F5 F6 F7 F8 F9 C1 C2 Levofl oxacin hemihydrate 100 100 100 100 100 100 100 100 100 100 100<br />

HPMC K100LV 100 100 100 150 150 150 200 200 200 125 175<br />

Sodium bicarbonate 40 60 80 40 60 80 40 60 80 50 70<br />

Magnesium stearate 4.8 5.2 5.6 5.8 6.2 6.6 6.8 7.2 7.6 5.5 6.9<br />

Talc 2.4 2.6 2.8 2.9 3.1 3.3 3.4 3.6 3.8 2.8 3.5<br />

*Weight expressed as mg per tablet; HPMC-hydroxypropyl methylcellulose; LV-low viscosity; C 1 , C 2 check point formulations.<br />

Duration of fl oating and fl oating lag time was measured<br />

by visual observation.<br />

Swelling index19 The tablet was weighed (W ) and placed in a glass<br />

Wet<br />

beaker, containing 200 mL of 0.1 N HCl, maintained in<br />

a water bath at 37 ± 0.5ºC. At regular time intervals, the<br />

tablet were removed and the excess surface liquid was<br />

carefully removed by a fi lter paper. The swollen tablet<br />

was then reweighed (W ). The swelling index (SI) was<br />

Dry<br />

calculated using the Equation 2.<br />

W − W<br />

Swelling Index =<br />

W<br />

In-vitro dissolution study<br />

wet Dry<br />

Dry<br />

×100 (2)<br />

In-vitro dissolution study of levofl oxacin hemihydrate<br />

fl oating tablets was carried out in USP XXIII tablet<br />

dissolution test apparatus (Electro lab TDT-08L),<br />

employing a paddle stirrer at 50 rpm using 900 ml of<br />

pH 1.2 solution as dissolution medium at 37 ± 0.5ºC.<br />

One tablet was used in each test. At predetermined<br />

time intervals, 5 ml of the samples were withdrawn by<br />

means of a syringe fi tted with pre-fi lter. The volume<br />

withdrawn at each interval was replaced with same<br />

quantity of fresh dissolution medium. The samples were<br />

analyzed for drug release by measuring the absorbance<br />

at 293 nm using Shimadzu UV-1800 spectrophotometer<br />

after suitable dilutions. All the studies were conducted<br />

in triplicate.<br />

Stability study<br />

Short-term stability study was performed at a temperature<br />

of 45° ± 1°C and 75% relative humidity over a period of<br />

thee weeks for the promising GRDF tablet formulation<br />

F . 15 tablets were packed in amber colored, screw-<br />

3<br />

capped bottles and kept in hot air oven maintained at<br />

45° ± 1°C and 75% relative humidity. Samples were taken<br />

at weekly interval for drug content estimation. At the end<br />

of three weeks, dissolution test and in-vitro fl oating study<br />

were performed to determine the drug release profi les,<br />

in-vitro fl oating lag time and fl oating time.<br />

RESULTS AND DISCUSSION<br />

The hardness of the gastroretentive fl oating tablets<br />

of levofl oxacin hemihydrate was found to be in the<br />

range of 6.45 to 6.74 kg/cm² for the factorial design<br />

formulations. The friability of all tablets was from 0.53<br />

to 0.89 % for the factorial design formulations. The<br />

percentage deviation from the mean weights of all the<br />

batches was found to be within the prescribed limits as<br />

per IP. The low values of standard deviation indicated<br />

uniform drug content (Table 2). The swelling index of<br />

the tablets was increased with an increase in the polymer<br />

content and the content of gas generating agent i.e.<br />

sodium bicarbonate (Table 2).<br />

In-vitro fl oating study was performed by placing tablets<br />

in USP XXIII tablet dissolution apparatus containing<br />

900 ml of pH 1.2 maintained at a temperature of<br />

37 ± 0.5ºC. The fl oating lag time and fl oating time were<br />

noted visually. The results were presented in Table 2.<br />

For factorial formulations, lag time was in the range of<br />

10.33 to 64.57 sec. With formulations containing the<br />

same amount of polymer of the same grade, fl oating<br />

lag time was decreased with increase in concentration of<br />

sodium bicarbonate. For formulation F 3 , it was lowest<br />

(10.33 sec) as the drug-polymer (HPMC K100LV) ratio<br />

is 1:1 and sodium bicarbonate is in highest proportion<br />

among the formulations and the tablet remained intact<br />

for 12 h, while for formulation F 7 , lag time was highest<br />

(64.57 sec) as drug-polymer ratio is 1:2 and sodium<br />

bicarbonate is in lowest proportion (40 mg). All the<br />

designed formulations displayed a fl oating time of more<br />

than 24 h.<br />

In-vitro drug release study was performed using USP<br />

XXIII tablet dissolution apparatus From the above data,<br />

it was evident that as the proportion of polymer in the<br />

formulation increased, cumulative percent drug release in<br />

12 h decreased and as the proportion of the gas generating<br />

agent increased, the drug release was increased (Table 3).<br />

Factorial design<br />

Based on the preliminary trial, the levels of independent<br />

variables (X and X ) were fi xed at 100, 150, 200 mg for<br />

1 2<br />

HPMC K100LV (X ) and at 40, 60, 80 mg for sodium<br />

1<br />

bicarbonate (X ) in designing the formulations of 3²<br />

2<br />

40 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


D.Nagendrakumar et al.: Design and Optimization of Levofl oxacin Gastroretentive Tablets<br />

Table 2: Evaluation Results of Factorial Design Formulations of Levofl oxacin Hemihydrate GRDDS<br />

Formulation Code<br />

Mean Hardness<br />

(kg/cm 2 ) Friability (%)<br />

Average<br />

Weight (mg)<br />

Mean Drug Content<br />

Percent* ± SD<br />

Swelling<br />

Index* ± SD<br />

Floating Lag<br />

Time (sec)<br />

Floating<br />

time (h)<br />

F 1 6.61 0.53 248.26 98.60 ± 1.51 4.25 ± 1.35 46.21 >24<br />

F 2 6.55 0.67 268.15 99.64 ± 1.74 13.26 ± 1.87 12.61 >24<br />

F 3 6.45 0.71 289.73 101.48 ± 0.97 49.82 ± 1.79 10.33 >24<br />

F 4 6.69 0.74 300.02 100.05 ± 0.42 07.38 ± 0.06 57.28 >24<br />

F 5 6.62 0.79 319.69 101.87 ± 1.68 23.49 ± 0.48 18.48 >24<br />

F 6 6.51 0.84 342.95 101.62 ± 1.57 53.18 ± 1.42 14.69 >24<br />

F 7 6.74 0.85 352.27 99.74 ± 0.38 10.75 ± 1.29 64.57 >24<br />

F 8 6.67 0.87 369.86 101.85 ± 1.35 28.37 ± 1.15 24.29 >24<br />

F 9 6.59 0.89 393.48 99.39 ± 1.06 56.24 ± 1.45 18.54 >24<br />

*Average of thee determinations, values shown in parenthesis are standard deviations. Formulation F 3 was selected as the best and used for further studies; GRDDSgastroretentive<br />

drug delivery system.<br />

Table 3: In- vitro Cumulative Percent Release of Levofl oxacin Hemihydrate for Factorial Design Batches<br />

Cumulative Percent Drug Release Mean* ± SD<br />

Time (h) F1 F2 F3 F4 F5 F6 F7 F8 F9 01 8.84 ± 0.29 10.37 ± 0.96 13.16 ± 0.65 8.31 ± 0.72 9.51 ± 0.49 09.88 ± 0.34 10.58 ± 0.10 7.51 ± 0.45 6.98 ± 0.17<br />

02 16.96 ± 0.00 18.89 ± 0.90 24.15 ± 0.17 13.24 ± 0.49 13.54 ± 0.89 15.57 ± 0.45 17.40 ± 1.09 14.90 ± 1.44 15.27 ± 1.17<br />

03 24.51 ± 0.50 26.48 ± 0.96 32.33 ± 0.40 20.76 ± 0.79 21.72 ± 1.00 26.13 ± 0.50 24.42 ± 1.17 21.22 ± 1.34 24.29 ± 2.43<br />

04 32.66 ± 2.05 34.06 ± 0.64 40.92 ± 0.72 29.44 ± 1.09 28.88 ± 1.09 36.87 ± 0.47 29.81 ± 2.05 31.91 ± 1.54 34.18 ± 0.61<br />

05 41.65 ± 1.54 42.44 ± 1.15 45.18 ± 0.42 38.36 ± 1.00 35.47 ± 1.12 43.05 ± 0.41 35.70 ± 3.71 39.10 ± 1.20 42.05 ± 0.44<br />

06 48.51 ± 1.41 49.50 ± 0.55 50.05 ± 0.20 46.65 ± 0.80 46.15 ± 0.60 50.13 ± 0.40 44.22 ± 2.75 49.09 ± 1.20 47.15 ± 0.80<br />

07 57.35 ± 1.79 58.25 ± 1.59 63.14 ± 0.56 53.00 ± 1.19 57.67 ± 1.80 54.54 ± 0.54 49.97 ± 1.84 52.58 ± 1.20 56.38 ± 0.61<br />

08 63.41 ± 0.47 65.00 ± 0.74 67.05 ± 1.13 62.39 ± 2.00 60.00 ± 0.37 60.39 ± 0.85 57.16 ± 2.34 56.06 ± 1.20 66.18 ± 0.86<br />

09 71.06 ± 2.42 72.13 ± 0.15 72.18 ± 0.85 66.68 ± 1.34 69.38 ± 1.21 73.51 ± 0.90 64.02 ± 2.05 63.05 ± 1.20 70.05 ± 1.65<br />

10 77.26 ± 2.80 85.61 ± 0.15 88.03 ± 1.02 73.47 ± 2.42 77.36 ± 1.35 79.30 ± 1.95 72.24 ± 2.57 78.54 ± 1.20 72.05 ± 1.14<br />

11 82.58 ± 3.05 91.8 ± 0.37 97.52 ± 0.26 76.78 ± 0.77 80.14 ± 1.43 82.13 ± 0.54 77.26 ± 2.71 81.13 ± 1.32 75.18 ± 1.77<br />

12 87.23 ± 2.67 96.49 ± 0.51 98.91 ± 0.20 83.05 ± 2.50 85.32 ± 1.88 88.91 ± 2.96 81.72 ± 2.22 83.58 ± 1.31 79.13 ± 1.81<br />

*Average of thee determinations, values shown in parenthesis are standard deviations.<br />

Table 4: Dissolution Parameters for 3² Full Factorial Design Batches<br />

Batch Code<br />

Variable Level in<br />

Coded Form<br />

* #<br />

X X 1<br />

2<br />

t 50% (h) t 70% (h)<br />

Cumulative Percent<br />

Drug Release in 12 h<br />

F1 –1 –1 6.20 8.91 87.23<br />

F2 –1 0 6.13 8.71 96.49<br />

F3 –1 1 5.95 8.52 98.91<br />

F4 0 –1 6.68 9.36 83.05<br />

F5 0 0 6.36 9.18 85.32<br />

F6 0 1 6.01 8.74 88.91<br />

F7 1 –1 7.16 9.72 81.72<br />

F8 1 0 6.27 9.47 83.58<br />

F9 1 1 6.32 8.93 79.13<br />

C1 -0.5 +0.5 6.21 8.94 90.98<br />

C2 +0.5 +0.5 6.56 9.23 85.31<br />

C 1 , C 2 check point batches; t 50% , t 70% analyzed by matrix model fi tting using PCP Disso V3 Software. * For HPMCK100LV cps (X 1 ); transformed levels in mg are: –1=100;<br />

‘0’=150, +1=200, –0.5=125, +0.5=175; # For NaHCO 3 (X 2 ), transformed levels in mg are: –1=40; ‘0’=60, +1=80, –0.5=50, +0.5=70; All the batches contained 100 mg of<br />

levofl oxacin hemihydrate,1% talc and 2% magnesium stearate.<br />

full factorial design. The dissolution parameters i.e.,<br />

t 50% , t 70% values were selected as dependent variables.<br />

The formulation codes of the nine batches of factorial<br />

formulations along with dissolution parameter values<br />

(t 50% , t 70% ) and cumulative percent drug released in 12 h<br />

were shown in (Table 4). From the data, it was evident<br />

that formulation F 3 showed highly satisfactory values<br />

for dissolution parameters (t 50% 5.95 h, and t 70% 8.52 h)<br />

and released approximately 98.91% drug in 12 h.<br />

Hence, formulation F 3 was considered as the optimized<br />

promising levofl oxacin hemihydrate gastric fl oating drug<br />

delivery system with improved bioavailability.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 41


Development of polynomial equations<br />

D.Nagendrakumar et al.: Design and Optimization of Levofl oxacin Gastroretentive Tablets<br />

From the data of dissolution parameters shown in<br />

Table 4 for factorial formulations F 1 to F 9 , polynomial<br />

equations for two dependent variables (t 50% , and t 70% )<br />

have been derived using PCP Disso 2000 V3 software.<br />

The general polynomial equation (Equation 3) for 3² full<br />

factorial design was<br />

2 2 Y = b +b X +b X +b X X +b X +b22X (3)<br />

0 1 1 2 2 12 1 2 11 1 2<br />

Where, Y is dependent variable, b arithmetic mean<br />

0<br />

response of nine batches, and b estimated coeffi cient<br />

1<br />

for factor X . The main effects (X and X ) represented<br />

1 1 2<br />

the average result of changing one factor at a time<br />

from its low to high value. The interaction term<br />

(X X ) showed how the response changes when two<br />

1 2<br />

factors were simultaneously changed. The polynomial<br />

2 2 term (X and X2 ) were included to investigate non-<br />

1<br />

linearity.<br />

The equation derived for t was Y = 6.28 + 0.216X 50% 1 1<br />

– 0.283 X . The negative sign for coeffi cient of X 2 2<br />

indicates that, as the concentration of gas generating<br />

agent (sodium bicarbonate) increases, t value<br />

50%<br />

decreases.<br />

The equation derived for t was Y = 9.02+ 0.31 X –<br />

70% 2 1<br />

0.30 X . 2<br />

The validity of the equations was verifi ed by designing<br />

two check point formulations (C and C ) and studying<br />

1 2<br />

the drug release profi les. The dissolution parameters<br />

predicted from the equations derived and those<br />

observed from experimental results were summarized in<br />

.<br />

Figure 2: Contour plot showing effect of factorial variables on t50% Figure 1: Response surface plot showing effect of factorial vari-<br />

.<br />

ables on t50% Table 5. The closeness of predicted and observed values<br />

for t 50% and t 70% indicated validity of derived equations<br />

for the dependent variables. The computer generated<br />

response surfaces and contour plots for the dependent<br />

variables were shown in Fig. 1–4.<br />

Stability study<br />

Short-term stability study was performed on the<br />

promising formulation F by storing the samples at<br />

3<br />

45 ± 1ºC for 3 weeks. The samples were tested for any<br />

changes in physical appearance and drug content at<br />

weekly intervals. Statistical analysis was performed on<br />

the drug content data and drug release parameters by<br />

using Student t-test. The t value for the drug content<br />

was found to be 3.85. For t and t the t- values<br />

50% 70%,<br />

were found to be 3.46 and 3.00, respectively (Table 6).<br />

These results indicated that there were no signifi cant<br />

changes in drug content and dissolution profi le of<br />

the formulation F during storage at 45ºC for 3 weeks<br />

3<br />

shown no signifi cant effect on physical characteristics<br />

and drug content.<br />

CONCLUSIONS<br />

It was concluded that the gastroretentive fl oating<br />

systems of levofl oxacin hemihydrate with shorter<br />

lag time was prepared by direct compression method<br />

using HPMC K100LV and sodium bicarbonate as<br />

gas generating agent. As the amount of polymer in<br />

the tablet formulation increased, the drug release rate<br />

decreased and as the concentration of gas generating<br />

agent (sodium bicarbonate) increased, the drug release<br />

increased and at the same time fl oating lag time decreased.<br />

42 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


D.Nagendrakumar et al.: Design and Optimization of Levofl oxacin Gastroretentive Tablets<br />

.<br />

Figure 3: Response surface plot showing effect of factorial variables on t70% .<br />

Figure 4: Contour plot showing effect of factorial variables on t70% Table 5: Dissolution Parameters Predicted and Observed<br />

Values for Check point batches<br />

Predicted<br />

Values (h)<br />

Observed<br />

Values (h)<br />

Formulations t50% t70% t50% t70% C1 6.31 9.01 6.20 8.90<br />

C2 6.20 9.02 6.50 9.20<br />

Table.6: Statistical Analysis of Dissolution Parameters<br />

(t 50% , t 70% ) of Stability study for Batch F 3<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 43<br />

Trial<br />

t 50% Values<br />

1st Day<br />

(A)<br />

21st<br />

Day<br />

(B)<br />

A-B<br />

t 70% Values<br />

1st<br />

Day<br />

(A)<br />

21st<br />

Day<br />

(B)<br />

A-B<br />

I 6.01 5.98 0.03 8.69 8.65 0.04<br />

II 5.94 5.93 0.01 8.74 8.73 0.01<br />

III 5.89 5.87 0.02 8.72 8.69 0.03<br />

Mean 5.95 5.92 0.02 8.71 8.69 0.026<br />

SD ± ± 0.060 ± 0.055 ± 0.01 ± 0.025 ± 0.04 ± 0.015<br />

t = 3.46 ;( p


REFERENCES<br />

D.Nagendrakumar et al.: Design and Optimization of Levofl oxacin Gastroretentive Tablets<br />

1. Prajapati ST, Patel LD, Patel DM. Gastric fl oating matrix tablets:<br />

Design and optimization using combination of polymers. Acta Pharm<br />

2008;58:221–9.<br />

2. Sauzet C, Claeys-Bruno M, Nicolas M, Kister J, Piccerelle P, Prinderre<br />

P. An innovative fl oating gastro retentive dosage system: Formulation<br />

and in-vitro evaluation. Int J Pharm 2009;378 (1–2):23–9.<br />

3. Garg R, Gupta GD. Progress in controlled gastroretentive delivery<br />

systems. Trop J Pharm Res 2008;7(3):1055–66.<br />

4. Xiaoqiang X, Minjie S, Feng Z, Yiqiao H. Floating matrix dosage form for<br />

phenoporlamine hydrochloride based on gas forming agent: in-vitro and<br />

in-vivo evaluation in healthy volunteers. Int J Pharm 2006;310:139–45.<br />

5. Deshpande AA, Shah NH, Rhodes CT, Malick W. Development of a<br />

novel controlled release system for gastric retention. Pharm Res 1997;<br />

14:815–9.<br />

6. Chavanpatil MD, Jain P, Chaudhari S, Shear R, Vavia RR. Novel<br />

sustained release, swellable and bioadhesive gastroretentive drug<br />

delivery system for ofl oxacin. Int J Pharm 2006;316 (1–2):86–92.<br />

7. Hwang SJ, Park H, Park K. Gastric retentive drug-delivery systems. Crit<br />

Rev Ther Drug Carrier Syst 1998;15 (3):243–84.<br />

8. Seth PR, Tossounian J. The hydrodynamically balanced system,<br />

a novel drug delivery system for oral use. Drug Dev Ind Pharm<br />

1984;10:313–9.<br />

9. Harrigan RM. Drug delivery device for preventing contact of undissolved<br />

drug with the stomach lining. US patent 4055178. 1977 October 25.<br />

10. Whitehead L, Fell JT, Collett JH. Development of a gastroretentive<br />

dosage form. Eur J Pharm Sci 1996;4(Suppl.):S182.<br />

11. Kawashima Y, Niwa T, Takeuchi H, Hino T, Itoh Y. Hollow microspheres<br />

for use as a fl oating controlled drug delivery system in the stomach.<br />

J Pharm Sci 1992;81:135–40.<br />

12. Chen J, Blevins WE, Park H, Park K. Gastric retention properties of<br />

superporous hydrogel composites. J Control Release 2000;64(1–3):<br />

39–51.<br />

13. Gröning R, Berntgen M, Georgarakis M. Acyclovir serum concentrations<br />

following peroral administration of magnetic depot tablets and the<br />

infl uence of extracoporal magnets to control gastrointestinal transit. Eur<br />

J Pharm Biopharm 1998;46:285–91.<br />

14. Singh BM, Kim KH. Floating drug delivery systems: An approach to<br />

oral controlled drug delivery via gastric retention. J Control Relese<br />

2000;63:235–59.<br />

15. Md. Ismail M, Dehghan MH, Shaikh A, Sahuji T, Chudiwal P. Preparation<br />

of a novel fl oating ring capsule-type dosage form for stomach specifi c<br />

delivery. Soudi Pharm J 2011;19:85–93.<br />

16. [updated 2011 January 6; cited 2011 July 17]. Available from: http://<br />

www.rxlist.com/Levaquin-drug.htm<br />

17. Lachman L, Liberman HA, Kanig JL. The theory and practice of<br />

industrial pharmacy. 3rd ed. Mumbai: Varghese Publishing House;<br />

1991. p. 297–303.<br />

18. Jimenez-Castellanos MR, Hossein Z, Rhodes CT. Design and testing<br />

in vitro of a bioadhesive fl oating drug delivery system for oral application.<br />

Int J Pharm 1994;105:65–70.<br />

19. Tadros IM. Controlled-release effervescent fl oating matrix tablets of<br />

ciprofl oxacin hydrochloride: Development, optimization and in vitro in<br />

vivo evaluation in healthy human volunteers. Eur J Pharm Biopharm<br />

2010;74:332–9.<br />

44 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Study of the binding properties of hydroxypropyl<br />

guar and its utilization in the formulation and<br />

evaluation of metoprolol tartarate tablets<br />

Swamy N.G.N.*, Dharmarajan T.S., Paranjothi K.L.K<br />

*Department of Pharmaceutics, Government College of Pharmacy, Bangalore – 560 027<br />

ABSTRACT<br />

Derivatization of guar to hydroxypropyl guar enhances the interaction coeffi cient and checks the hydration<br />

rate of the polymer molecule. Hydroxypropyl guar is investigated for its binding property along with sodium<br />

carboxymethylcellulose (1% w/w) as the parallel binding agent. Tablets of metoprolol tartarate were prepared<br />

by wet granulation. Pre-compression parameters such as rate of fl ow, angle of repose, Carr’s index, Hausner’s<br />

ratio, residual moisture content and assay determinations were carried out. Hydroxypropyl guar bound granules<br />

revealed lower Carr’s index value and Hausner’s ratio in comparison to sodium carboxymethylcellulose bound<br />

granules thereby indicating better free fl owing properties. In respect to post compression parameters, tablets<br />

made with hydroxypropyl guar displayed superior hardness, lesser deviation of tablet weight about the mean, a<br />

higher value of (Crushing strength: Friability)/Disintegration time, a faster drug dissolution in comparison to sodium<br />

carboxymethylcellulose bound tablets. While guar with its high intrinsic viscosity restricts its use as a retardant<br />

component in matrix tablets, hydroxypropyl guar with its enhanced interaction coeffi cient and controlled hydration<br />

rate has yielded a promising adjuvant for binding metoprolol tartarate tablets with improved in vitro performance.<br />

Keywords: Carr’s index, crushing strength, disintegration time, friability, hausner’s ratio, hydroxypropyl guar,<br />

interaction coeffi cient, rate of hydration, residual moisture content.<br />

INTRODUCTION<br />

Guar gum1 consists of the endosperm part<br />

of guar plant namely Cyamopsis tetragonolobus<br />

Linn. (Family: Leguminosae). When used in<br />

extemporaneous preparations, guar imparts<br />

colour, which is not desirable. Guar has a<br />

very high intrinsic viscosity but very poor<br />

interaction coeffi cient. 2 Derivatization of<br />

guar to hydroxypropyl guar improves its<br />

interaction coeffi cient. 3 By virtue of its<br />

high intrinsic viscocity, guar gum has been<br />

extensively used as a retarding component<br />

in the formulation of matrix tablets. 4–18<br />

Hydroxypropyl guar with improved<br />

interaction coeffi cient has been investigated<br />

as a gelling agent, 19 as a bodying agent20 in<br />

suppositories, fi lm forming agent21 and<br />

as a suspending agent. 22 Hydroxypropyl<br />

guar with a controlled rate of hydration<br />

is investigated for its solution binding<br />

properties 23 in the formulation of oral<br />

tablets. Sodium carboxymethylcellulose is<br />

tried as the parallel binding agent. 24–28<br />

Metoprolol tartarate 29 (MT), is a selective<br />

β-adrenoreceptor antagonist useful in<br />

the management of hypertension and<br />

ischaemic heart diseases. MT 30 is readily<br />

absorbed after oral administration and<br />

rapidly distributed into body tissues.<br />

The elimination half-life is about 3–7 h.<br />

The drug undergoes extensive fi rstpass<br />

metabolism; only 50% of the orally<br />

administered drug reaches the systemic<br />

circulation. Adult oral dose is 50 mg twice<br />

daily to be optimized subsequently and<br />

not to exceed 400 mg per day. Tablets<br />

containing 50 mg of MT per tablet were<br />

formulated using hydroxypropyl guar<br />

Research Ar cle<br />

Received Date : 26-12-2011<br />

Revised Date : 01-03-2012<br />

Accepted Date : 05-03-2012<br />

DOI: 10.5530/rjps.2012.1.6<br />

Address for<br />

correspondence<br />

Dr. N.G.N. Swamy<br />

Professor<br />

Department of Pharmaceutics<br />

Government College of<br />

Pharmacy,<br />

No. 2, P. Kalinga Rao Road,<br />

Bangalore – 560 027<br />

Mob. No: + 91 9945451772<br />

E-mail: ngnswami@yahoo.co.in<br />

www.rjps.in<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 45


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

(HPG) and sodium carboxymethylcellulose (Na CMC)<br />

as binders.<br />

MATERIALS AND METHODS<br />

MT I.P. was obtained as a gift sample from ASTRA-<br />

IDL ltd. Na CMC (high viscosity grade), Lobachemie<br />

make was used. All other excipients used were of Pharmacopoeial<br />

grade. HPG was obtained by derivatization<br />

of guar, 3 MT tablets weighing around 150 mg each and<br />

containing 50 mg of the drug were made by wet granulation<br />

by employing 1% W/V aqueous dispersions<br />

of HPG and Na CMC respectively as solution binders.<br />

1% W/V HPG dispersion revealed a viscosity of<br />

6400 cps; Na CMC displayed a viscosity of 448 cps.<br />

The aqueous polymer dispersions were stabilized with<br />

0.18% W/V methyl paraben and 0.02% W/V propyl<br />

paraben.<br />

The formulation details of the tablets are shown in<br />

Table 1.<br />

PART-I: PREPARATION OF GRANULES<br />

The quantity of MT as specifi ed in Table 1 was mixed<br />

with lactose IP in the ascending order of weights; the<br />

granulating agent was added little by little so as to get<br />

a damp mass with a pliable consistency. The batter was<br />

allowed to stand for 15 min and passed through sieve<br />

20; the granules so obtained were dried in an oven at 60°<br />

for 1 h duration. The agglomerates were regranulated by<br />

using sieve 20 superimposed on sieve 40. While granules<br />

retained on sieve 20 were used for compression, the<br />

fi nes retained on sieve 40 were used for improvement<br />

of fl ow properties of granules.<br />

PART-II: STUDY OF GRANULE PROPERTIES<br />

(a) Determination of compressibility factor<br />

The untapped and the tapped bulk densities were<br />

obtained by the 10 point method. The 10 cc tared<br />

measuring jar was fi lled with granules up to 1 cc and<br />

the weight was recorded; likewise, the weight of 2 cc,<br />

3 cc ... up to 10 cc was fi lled with granules and the weight<br />

was recorded without tapping. Similarly the procedure<br />

was repeated with the granules fi lled up to 1 cc, tapped<br />

so as to compact the granules, excess granules was added<br />

to record a volume of 1 cc after tapping. The weight<br />

of 2 cc, 3 cc ... up to 10 cc with tapping each time after<br />

the addition of excess granules to adjust the volume<br />

was recorded. A graph of weight values (gm) vs corresponding<br />

volume (cc) was constructed. The bulk density<br />

was arrived from the slope of the linear relationship.<br />

Compressibility factor ‘C’ was calculated using the formula,<br />

c<br />

b a<br />

=<br />

b<br />

− (1)<br />

Table 1: The formulation details of Metoprolol tartarate<br />

tablets<br />

Tablet wt Tablet wt<br />

Ingredient<br />

in gm in gm<br />

Metoprolol tartarate 30 30<br />

Lactose I.P 60 60<br />

Hydroxypropyl guar 1% w/v aqueous<br />

dispersion<br />

Q.S –<br />

Sodium CMC 1% w/v aqueous<br />

dispersion<br />

– Q.S<br />

Talc 2% 2%<br />

Where, b=Tapped bulk density and a = Untapped bulk<br />

density. The graphical representations are shown in<br />

fi gure 1and 2. Compressibility factor multiplied by 100<br />

gives Carr’s index values.<br />

(b) Determination of the optimum amount of<br />

Lactose fi nes to be incorporated to the 20/40<br />

mesh granules to improve the fl ow properties<br />

Since 1% w/v HPG bound granules revealed the lowest<br />

Carr’s index value, 31 1% w/v HPG bound lactose<br />

granules were used for the evaluation.<br />

(i) Determination of angle of repose32 Carried out by employing fi xed bed method using the<br />

formula,<br />

tan θ = ;<br />

h<br />

(2)<br />

r<br />

Where, θ = angle of repose, h = height of the heap<br />

and r = radius of the heap. The exact value of θ was<br />

arrived at by referring to the table on natural tangents.<br />

The advantage of this method being, the dimension of<br />

the base is fi xed, whereas, the height alters as the angle<br />

of repose changes.<br />

(ii) Determination of the rate of fl ow33 Carried out using a glass funnel having an orifi ce and<br />

base diameters of 0.8 cm and 9 cm respectively and<br />

securely clamped to a retort stand. A 50 gm sample was<br />

introduced into the funnel and the powder was allowed<br />

to fall freely under the gravitational force. The fl ow rate<br />

was calculated using the expression:<br />

Flow rate = amount of powder (gm) / time of fl ow (s). (3)<br />

The above studies were conducted on 20/40 # lactose<br />

granules blended with 5% w/w, 10% w/w and 15%<br />

w/w, lactose fi nes. The average value of three such<br />

determinations is recorded in fi gures 3 and 4.<br />

(c) Determination of optimum concentration of<br />

the glidant (Talc) to be added to dummy lactose<br />

granules containing 10% w/w of Lactose fi nes<br />

The θ values and the fl ow rate values were determined<br />

for the above mentioned granules blended with 1%, 2%,<br />

3%, 4% and 5% w/w talc powder. The infl uence of the<br />

46 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

Figure 1: Untapped and tapped BD recording for 20/40#MT granules made with 1% w/v of HPG.<br />

Figure 2: Untapped and tapped BD recording for 20/40#MT granules made with 1% w/v of NaCMC.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 47


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

Figure 3: Influence of percentage lactose fines on the angle of repose for 20/40# lactose granules.<br />

Figure 4: Influence of percentage lactose fines on the rate of flow for 20/40# lactose granules.<br />

talc % w/w on the θ and the fl ow rates is depicted in<br />

fi gure 5 and 6.<br />

From the studies carried out at “b” and “c” above, it<br />

could be concluded that the optimum fi nes concentration<br />

was 10% w/w and optimum glidant concentration was<br />

2% w/w of talc powder.<br />

(d) Determination of Carr’s index for 20/40 mesh<br />

MT granules containing 10% w/w fi nes and<br />

blended with 2% w/w of Talc powder<br />

The graphical representation for granules made with<br />

1% w/v HPG as binder are shown in fi gure 7 and<br />

for granules made with 1% w/v NaCMC is shown<br />

in fi gure 8. Carr’s Index is calculated by using the<br />

formula<br />

CI = (Dt – Db/ Dt) × 100 (4)<br />

(e) Hausner’s ratio<br />

It is an index of ease for powder fl ow. It is calculated by<br />

the following formula,<br />

Hausner s ratio Dt<br />

' = (5)<br />

Db<br />

48 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

Figure 5: Influence of % w/w talc added on the angle of repose of 20/40 mesh lactose granules blended with 10% w/w of lactose fines.<br />

Figure 6: Influence of % w/w talc added on the flow rate of 20/40 mesh lactose granules blended with 10% w/w of lactose fines.<br />

Table-2: Infl uence of additives on the angle of repose and the fl ow rate for dummy 20/40 # Lactose granules<br />

Granules composition<br />

20/40 mesh lactose<br />

granules bound with<br />

1% w/v HPG,<br />

20/40 mesh lactose<br />

granules bound with<br />

1%w/v NaCMC,<br />

Angle of<br />

repose for<br />

plain granules<br />

Angle of repose<br />

with the added<br />

additives<br />

Fall in angle<br />

of repose<br />

Flow rate in gm/s<br />

for granules with<br />

additives<br />

Flow rate in<br />

gm/s for plain<br />

granules<br />

Enhancement<br />

in fl ow rate<br />

(gm/s)<br />

40°7’ 38°42’ 1°25’ 3.508 3.385 0.123<br />

39°11’ 37°18’ 1°53’ 3.533 3.496 0.037<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 49


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

Weight (gm)<br />

Figure 7: Untapped and tapped BD recording for 20/40# MT granules made with 1% w/v HPG blended with 10% w/w fines and 2% w/w talc.<br />

Where Dt is the tapped bulk density and Db is the<br />

untapped bulk density.<br />

(f) Determination of angle of repose θ<br />

Angle of repose determination and fl ow time<br />

determination were carried out on 20/40 mesh dummy<br />

granules blended with 10% w/w lactose and 2% W/W<br />

talc powder.<br />

The infl uence of additives on the fl ow properties of<br />

20/40 mesh dummy granules is depicted in Table 2<br />

(g) Determination of residual moisture content<br />

Residual moisture content was determined by drying the<br />

granules at 60° for 15 min. 34<br />

(h) Assay of granules<br />

(i) Carried out to ascertain uniform distribution of drug<br />

in tablets over the entire range of sample (ii) To enable<br />

us to adjust the die volume/ the total weight of the tablet<br />

so as to get tablets having the required drug content. MT<br />

has a λmax at 275 nm in 95% v/v ethanol. It obeys Beer’s<br />

Lamberts law in the concentration range of 10–100<br />

μg/ml. A calibration curve was constructed; the linear<br />

relationship had a slope value of 0.0046 and an intercept<br />

value of 0.0034 which were used in the calculation<br />

of the drug content in the assay of sample. The precompression<br />

parameters are contained in Table 3.<br />

PART-III: COMPRESSION OF GRANULES<br />

INTO TABLETS<br />

The compression was carried out using a 10 station<br />

rotary tablet machine. The capacity of the die cavity was<br />

so adjusted as to accommodate a weight of granules<br />

containing 50 mg of MT. 7mm punches were used and<br />

a compression pressure of 6 Kg was applied on the<br />

rollers for all tablets. The tablets were preserved in tight<br />

screw capped bottles until further evaluation tests were<br />

carried out.<br />

PART-IV: EVALUATION OF TABLETS<br />

The compressed tablets were subjected to the following<br />

evaluation tests.<br />

(i) Hardness test 35<br />

Carried out using Monsonto hardness tester; a mean of<br />

three readings was recorded.<br />

(ii) Determination of disintegration time36 Disintegration time was recorded as per I.P procedure;<br />

6 tablets were subjected to the study.<br />

(iii) Weight variation 36<br />

The test was carried out on 20 tablets. The tablets were<br />

weighed collectively and the average weight was arrived<br />

50 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

Table 3: Pre-compression parameters<br />

Granule composition<br />

20/40 mesh MT granules bound with<br />

1% w/v HPG, blended with 10% w/w fi nes,<br />

2% w/w talc.<br />

20/40 mesh MTgranules bound with<br />

1%w/v NaCMC, blended with 10% w/w<br />

fi nes, 2% w/w talc.<br />

RMC* Residual moisture content.<br />

Weight (gm)<br />

Untapped bulk<br />

density (Db) gm/cc<br />

at. The weights of 20 tablets were recorded individually<br />

and the deviation from the mean was calculated. Not<br />

more than 2 tablets to deviate beyond ±7.5% and not<br />

even one to deviate beyond ±15%.<br />

(iv) Friability Test 37<br />

The test was carried out on 20 tablets weighed collectively.<br />

The tablets were loaded on to a Roche friabilator and<br />

Tapped bulk<br />

density (Dt) gm/cc<br />

Carr’s<br />

Index<br />

0.5638 0.5761 2.13<br />

Hausner’s<br />

Ratio<br />

1.0218<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 51<br />

RMC*<br />

Assay Value<br />

(mg)<br />

1.34 47.51<br />

0.5346 0.5728 6.66 1.0714 1.24 48.71<br />

Volume (cc)<br />

Figure 8: Untapped and tapped BD recording for 20/40# MT granules made with 1%w/v NaCMC blended with 10% w/w fines and 2%w/w talc .<br />

Table 4: Post compression parameters<br />

Hardness<br />

in Kg/cm2 Disintegration Friability Assay<br />

time in minutes %<br />

(mg) (CS:FR)/DT* t in min<br />

80%<br />

Tablets made with 1% w/w<br />

HPG as binder<br />

4.5 8.20 0.24 47.86 2.3 20<br />

Tablets made with 1% w/v<br />

of NaCMC as Binder<br />

3.0 7.50 0.18 47.45 2.1 35.45<br />

(CS:FR)/DT*; CS = Crushing strength, FR = Friability and DT = Disintegration time.<br />

rotated at 25 rpm for 4 min; tablets weighed again and<br />

% friability was calculated. It is desirable that the weight<br />

loss is less than 1.0%.<br />

(v) Assay<br />

As mentioned under granules; 5 tablets36 are randomly<br />

powdered; a quantity equivalent to 100 mg of MT was<br />

taken and dissolved in 95% of ethanol; subsequent


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

Figure 9: Dissolution profile of 1% HPG and 1% Na CMC bound Metoprolol Tartarate tablets.<br />

dilutions were made with ethanol to have MT<br />

concentration in the range of 10–100 μg/ml; absorbance<br />

was read at 275 nm; concentration of MT was calculated<br />

by making use of slope and intercept values obtained<br />

from the calibration curve. Post compression parameters<br />

are compiled in Table 4.<br />

(vi) Dissolution profi le<br />

Dissolution testing was carried out in 0.1N Hydrochloric<br />

acid. Amount of medium used was 1000 ml; rpm 50.<br />

Volume withdrawn was 10 ml, samples withdrawn at<br />

the end of 15, 30, 45, 60, 75 and 90 min and analyzed<br />

spectrophometrically at 275 nm. The dissolution profi les<br />

of HPG bound and Na CMC bound tablets are depicted<br />

in fi gure 9.<br />

RESULT AND DISCUSSION<br />

In this study, 1% w/v aqueous dispersion of HPG<br />

has been tried as a binder for MT; 1%w/v aqueous<br />

dispersion of Na CMC is used as the parallel binder.<br />

20/40 mesh lactose granules were made and mixed<br />

with optimum amounts of 10% w/w lactose fi nes<br />

and 2% w/w of talc powder. 20/40 mesh granules as<br />

mentioned afore said were mixed with 10% w/w fi nes<br />

and 2%w/w talc powder and subjected to untapped and<br />

tapped density determination; from this the percentage<br />

compressibility factor i.e. Carr’s index and Hausner’s<br />

ratio were calculated. Granules made with HPG as<br />

binder revealed a Carr’s index value of 2.13 in contrast<br />

to a value of 6.66 for granules made with 1% w/v Na<br />

CMC dispersion. Granules having Carr’s index value in<br />

the range of 5 to 12 reveal excellent fl ow characteristics.<br />

Granules made with HPG revealed a Hausner’s ratio of<br />

1.022 in contrast to a value of 1.071 in case of Na CMC<br />

granules. Lower Hausner’s ratio (1.25). It is a well verifi ed fact<br />

that the more compressible a bed of particles is, the less<br />

fl owable the powder or granules will be. A material having<br />

a compressibility value of less than 16 is considered to<br />

be a free fl owing material; this is in close agreement with<br />

the data compiled in table 3. Repose angle also needs<br />

to be considered along with compressibility; Repose<br />

angle 34 for the experimental samples are in the range of<br />

37–38° which is marginally higher than the optimum<br />

value i.e. 30°. When the angle of repose exceeds 50°,<br />

the fl ow is rarely acceptable for manufacturing process.<br />

Table 5: Flow properties and corresponding angle of<br />

repose<br />

Flow property Angle of repose (Degrees)<br />

Excellent 25–30<br />

Good 31–35<br />

Fair 36–40<br />

Passable 41–45<br />

Poor 46–55<br />

Very poor 56–65<br />

Very, Very Poor >66<br />

52 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

The infl uence of angle of repose 38 on fl ow properties<br />

of granules is depicted in Table 5.<br />

The fl ow rate was determined by passing 50 gm of<br />

granules through a glass funnel with orifi ce and base<br />

diameters of 0.8 cm and 9 cm respectively. By and large,<br />

lower the compressibility factor and smaller the repose<br />

angle, the faster is the predicted fl ow rate of granules.<br />

HPG granules have revealed a signifi cant enhancement<br />

in the fl ow rate after the inclusion of additives i.e.<br />

0.123 gm/s in contrast to an enhancement value of<br />

0.037 gm/s incase of granules made with Na CMC.<br />

For trouble free compression, a moisture content of<br />

around 1.5% is desirable. HPG bound granules revealed<br />

a moisture content of 1.34% in contrast to a value of<br />

1.24 in case of granules made with Na CMC as binder.<br />

Moisture content of less than 1% can cause capping and<br />

lamination whereas above 2%, some sticking problem<br />

may develop.<br />

In respect to post compression parameters, from the<br />

weight variation data, it is observed that in case of HPG<br />

bound tablets, the % deviation from the mean was found<br />

to range between (-) 5.86 to (+) 6.73 in contrast to a<br />

value of (-) 7.39 to (+) 8.94 for Na CMC bound tablets.<br />

This observation correlates well with the lower ‘C’<br />

values obtained for HPG bound granules. Tablets made<br />

with HPG as binder revealed a hardness of 4.5 kg/cm 2<br />

in contrast to a value of 3 Kg/cm 2 for tablets made<br />

with Na CMC as binder. The friability value for HPG<br />

bound tablets was 0.24% in contrast to a value of 0.18%<br />

in case of Na CMC bound tablets. Disintegration time<br />

for HPG bound tablets was 8.3 min which was slightly<br />

higher than the Disintegration time value of 7.8 min for<br />

Na CMC bound tablets. Crushing strength – friability<br />

ratio (CS:FR) which is the quotient of the crushing<br />

strength (CS) value divided by the friability (FR) value, is<br />

an index used as a measure of the mechanical strength<br />

of tablets. But the (CS:FR)/DT ratio would be a better<br />

index for measuring tablet quality. 39 It is so because, in<br />

addition to measuring tablet strength (crushing), and<br />

tablet weakness (friability), it does evaluate all negative<br />

effects of these parameters on disintegration time. 40<br />

HPG bound tablets revealed a (CS:FR)/DT ratio of<br />

2.26 in contrast to a value of 2.1 in case of Na CMC<br />

bound tablets. A higher value of (CS:FR)/DT is<br />

indicative of a better balancing act between the cohesive<br />

binding and disintegrating behavior of the tablets. From<br />

the dissolution profi le, a t 80% value of 20 min has been<br />

displayed in case of HPG bound tablets in contrast to a<br />

value of 35.45 min in Case of Na CMC bound tablets.<br />

CONCLUSION<br />

The hydroxypropyl guar with it’s enhanced interaction<br />

coeffi cient and controlled rate of hydration has proved<br />

to be a superior solution binder in achieving suffi ciently<br />

rapid release of the drug from the tablet formulation in<br />

comparison to the conventionally used tablet solution<br />

binder namely sodium carboxymethylcellulose.<br />

ACKNOWLEDGEMENT<br />

The authors wish to thank Shri P. Guruswamy, The<br />

then plant manager, Caryll Pharma, Sarakki main road<br />

Bengaluru, for providing tablet compression facility;<br />

Shri K.R.P Shenoy, Controller, QA and Technical<br />

services, ASTRA-IDL for gifting Metoprolol Tartarate<br />

and Shri Chandrashekhar, Managing Director, Ce-Chem<br />

Pharmaceuticals Pvt Ltd. Peenya, Bengaluru for the gift<br />

Sample of Lactose I.P.<br />

REFERENCES<br />

1. Roy-L-Whistler. Industrial Gums Polysaccharides and their derivatives.<br />

New York: 2nd ed. Academic press;1973.<br />

2. Misra AN, Baveja JM. Modifi ed Guar gum as hydrophilic matrix for<br />

controlled release tablets. Indian Drugs 1997;34(4):216–23.<br />

3. Swamy NGN, Dharmarajan TS, Paranjothi KLK. Derivatization of guar<br />

to various hydroxyalkyl derivatives and their characterization. Indian<br />

Drugs 2006;43(9):756–9.<br />

4. Bhalla HL, Ghulati SO. Sustained release Theophylline tablets. Indian<br />

drugs 1987;24(7):338–42.<br />

5. Bhalla HL, Sanzgiri YD. An improvised controlled release tablets of<br />

Salbutamol sulphate. Indian J Pharm Sci 1987;49(1):22–5.<br />

6. Khullar P, Khar RK, Agarwal SP. Evaluation of Guar gum in the<br />

preparation of sustained release matrix tablets. Drug Dev Ind Pharm<br />

1998;24(11):1095–9.<br />

7. Altaf SA, Yu K, Parasrampuria J, Friend Dr. Guar gum based sustained<br />

release Diltiazem. Pharma Res 1998;15(8):1196–201.<br />

8. Krishnaih YSR, Satyanarayana S, Ramprasad YV, Narsimha Rao S.<br />

Gamma scientigraphic studies on guar gum matrix tablets for colonic<br />

drug delivery in healthy volunteers. J Controlled Release 1998;55<br />

(2–3):245–52.<br />

9. Krishnaih YSR, Veer Raju P, Dinesh Kumar B, Bhaskar P. Satyanarayana<br />

V. Development of colon targeted drug delivery systems of Mebendazole.<br />

J Controlled Release 2001;77(1–2):87–95.<br />

10. Sinha VR, Rachna Kumria. Polysaccharides in colon-specifi c drug<br />

delivery. Int J Pharm 2001;224(1–2):19–38.<br />

11. Krishnaih YSR, Latha K, Nageshwar RL, Karthikeyan RS, Bhaskar<br />

P, Satyanarayana V. Development of colon targeted oral guar gum<br />

matrix tablets of Albendazole for the treatment of Helminthiasis. Indian<br />

J Pharm Sci 2003;65(4):378–85.<br />

12. Hazare AA, More HN, Desouza JI. Design and evaluation of<br />

sustained release tablets of Diltiazem hydrochloride, Indian Drugs<br />

2004;41(3):175–6.<br />

13. Toti US, Tejaraj M, Aminabhavi. Modifi ed guar gum matrix tablet for<br />

controlled release of Diltiazem Hydrochloride. J Controlled Release<br />

2004;95(3):567–77.<br />

14. Al-Saidan SM, Krishnaih YSR, Patrao SS, Satyanarayana V. In-vitro<br />

and In-vivo evaluation of guar gum matrix tablets for oral controlled<br />

release of water soluble Diltiazem hydrochloride. AAPS Pharm Sci Tech<br />

2005;6(1):Article 5:E14-E21.<br />

15. Mishra B, Bansal A, Sankar C. Development and In-Vitro evaluation of<br />

hydrophilic matrix tablets of Diltiazem hydrochloride. Acta Pharma Turc<br />

2005;47:115–26.<br />

16. Varshosaz J, Tavakoli N, Kheirolahi F. Use of hydrophilic natural gums<br />

in the formulation of sustained release matrix tablets of Tramadol<br />

hydrochloride. AAPS Pharm Sci Tech 2006;7(1):Article-24:E1-E7.<br />

17. Jain S, Yadav SK, Patil UK. Preparation and evaluation of sustained<br />

release matrix tablets of Furosemide using natural polymers. Research<br />

J Pharm and Tech 2008; 1(4):374–6.<br />

18. Amit S Yadav, et al., Design and evaluation of Guar gum based<br />

controlled release matrix tablets of Zidovudine. J Pharm Sci Tech<br />

2010;2(3):156–62.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 53


Swamy NGN et al.: Study of the binding properties of hydroxypropyl guar and its utilization in the formulation and evaluation of metoprolol tartarate tablets<br />

19. Swamy NGN, Dharmarajan TS, Paranjothi KLK. Study of Hydroxypropyl<br />

guar for its gelling property and its use in the formulation of Tenoxicam<br />

gels. Pak J Pharm Sci 2007;20(1):61–6.<br />

20. Swamy NGN, Dharmarajan TS, Paranjothi KLK. Study of<br />

thickening properties of Hydroxypropyl guar and its use in the<br />

formulation and evaluation of Ibuprofen suppositories. Indian Drugs<br />

2007;44(12):925–9.<br />

21. Swamy NGN, Dharmarajan TS, Paranjothi KLK. Study of fi lm forming<br />

properties of Hydroxypropyl guar and its use in the preparation of<br />

medicated transdermal patches. Ind J Pharm Edu Res 2008;42(2):<br />

147–53.<br />

22. Swamy NGN, Dharmarajan TS, and Paranjothi KLK. Study of the<br />

rheological behavior of hydroxypropyl guar and its use in the formulation<br />

and evaluation of Sulphamethoxazole suspensions. Indian Drugs<br />

2009;46(12):25–39.<br />

23. Krycer I, Pope DG, Hersey JA. An evaluation of tablet binding agents<br />

Part-I solution binders. Powder Technol1983;34(1):39–51.<br />

24. Baveja SK, Rangarao KV, Arora J. Examination of natural gums and<br />

mucilages sustaining materials in tablet dosage forms. Indian J Pharm<br />

Sci 1988;50(2):89–92.<br />

25. Baveja SK, Rangarao KV, Arora J. Examination of natural gums and<br />

mucilages as sustaining materials in tablet dosage forms Part-II. Indian<br />

J Pharm Sci 1989;51(4):115–8.<br />

26. Ofoefule SI and Chukwar A. Application of Abelmoschus esculentus<br />

gums as a minimatrix for Furosemide and Diclofencac Sodium tablets.<br />

Indian J Pharm Sci 2001;63(6):532–5.<br />

27. Salunkhe KS, Kulkarni MV. Formulation and in-vitro evaluation of<br />

Dextrin matrix tablets of Ibuprofen for colon specifi c drug delivery. Pak<br />

J Pharm Sci 2008 Jan;21(1):17–20.<br />

28. Shendge RS, Sayyad FJ, Salunkhe KS, Bhalke RD. Development of<br />

colon specifi c drug delivery of Aceclofenac by using effective binder<br />

system of ethyl cellulose. Int J Pharm Bio Sci 2010;1(3): 1–5.<br />

29. Plosker GL, Clissold SP. Controlled release Metoprolol formulations;<br />

A review of their pharmacodynamics and pharmacokinetic properties<br />

and therapeutic use in hypertension and ischaemic heart diseases.<br />

Drugs 1992;43(3):382–414.<br />

30. Florey K. Analytical profi les of drug substances. Vol.12. London:<br />

Academic press; 325–7.<br />

31. Bhowmik D, Chiranjib B, Krishnakanth, Pankaj, Chandira RM. Fast<br />

dissolving tablets; an overview, JOCPR 2009;1(1):163–77.<br />

32. Hegde RP, Rhengold JL, Welch S, Rhodes CT. Studies of powder fl ow<br />

using recording powder fl ow meter and measurement of the dynamic<br />

angle of repose. J Pharm Sci 1985 Jan;74(1):11–15.<br />

33. Onunkwo GC. Evaluation of Okro gum as a binder in the formulation<br />

of Thiamine hydrochloride granules and Tablets. Research in<br />

Pharmaceutical Biotechnology 2010; 2(3): 33–9.<br />

34. Sanyal SN, Datta AK. Studies on the granules parameters of<br />

Paracetamol tablets, The Eastrern Pharmacist 1992;35(409):175–7.<br />

35. Reddy VP, Suneetha A. Release rate of Diclofenac sodium from different<br />

polymers. Ind J Pharm Sci 2000;62(4):313–6.<br />

36. Indian Pharmacopoeia vol-1. Ghaziabad, Published by Indian<br />

Pharmacopoeial Commission. 2007:182.<br />

37. European Pharmacopoeia, 5th ed. Vol-1. France, European Directorate<br />

for the Quality of Medicines of the council of Europe; 2005. 234.<br />

38. Japanese Pharmacopoeia, 15th ed. Tokyo, The stationery Offi ce,<br />

Society of Japanese Pharmacopoeia; 2007. 1730.<br />

39. Alebiowu G, Itiola OA. The infl uence of Pregelatinized starch disintegrant<br />

on interacting variables that act on disintegrant properties. Pharm Tech<br />

2003;27 August issue:28–32.<br />

40. Autamashih M, Isah AB, Allagh TS, Ibrahim MA. Use of anhydrous<br />

calcium phosphate and selected binders in the tablet formulation of<br />

a deliquescent crude plant extract: Vernonia galamensis (Asteraceae).<br />

JAPS 2011;1(8):118–22.<br />

54 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Phytochemical investigation of root extract<br />

of the plant Carissa spinarum<br />

Karunakar Hegde 1 , D. Satyanarayana 2 , Arun B. Joshi 3<br />

1 Department of Pharmacology, Srinivas College of Pharmacy, Valachil, Post- Parangepete, Mangalore- 574 143,<br />

Karnataka, India<br />

2Department of Pharmachemistry, N G S M Institute of Pharmaceutical Sciences, Mangalore- 574 160, Karnataka, India<br />

3Department of Pharmacognosy, Goa College of Pharmacy, Panaji- 403 001, Goa, India<br />

ABSTRACT<br />

From the petroleum ether extract of the roots of Carissa spinarum Linn. (Apocynaceae), six compounds namely<br />

stigmasterol, ursolic acid, lupeol, campesterol, 17-hydroxy-11-oxo-nor-β-amyrone and urs-12-ene-3β, 22β-diol-<br />

17-carboxylic acid have been isolated by column chromatography. Their structures were characterized by m.p.,<br />

IR, 1HNMR, 13CNMR and mass spectral data. However, the compounds stigmasterol, campesterol, 17-hydroxy-<br />

11-oxo-nor-β-amyrone and urs-12-ene-3β, 22β-diol-17-carboxylic acid were reported for the fi rst time from the<br />

root of this plant.<br />

Keywords: Carissa spinarum, Root extract, Sterol, Triterpenoids<br />

INTRODUCTION<br />

Carissa spinarum Linn. (Carissa opaca Stapf<br />

ex Haines) is a evergreen shrub with short<br />

stem and strong thorns in pairs, belonging<br />

to family Apocynaceae. The plant is widely<br />

distributed throughout the dry, sandy and<br />

rocky soils of India, Ceylon, Myanmar and<br />

Thailand. 1 In traditional system of medicine<br />

the plant is used as purgative, for the<br />

treatment of rheumatism, cleaning worm<br />

infested wounds of animals and in snake<br />

bite. 1,2 Earlier studies have shown that the<br />

extract of the plant possesses cardiotonic, 3<br />

anticonvulsant, 4 hepatoprotective, 5 antiarthritic,<br />

6 antibacterial, 7 potent antioxidant8 and CNS depressant activitiy. 9 Various<br />

cardiac glycosides, 10 caffeic acid, 11 ursolic<br />

acid, naringin12 germacrane sesquiterpene<br />

and lignans8 were reported from this plant.<br />

Since the chemical investigation of the<br />

roots of Carissa spinarum has not been dealt<br />

in detail, in the present study an effort was<br />

made to establish and report the chemical<br />

investigations, of the petroleum ether<br />

extract of the roots of this plant.<br />

MATERIALS AND METHODS<br />

Chemicals and Instruments<br />

All the chemicals and solvents were<br />

of analytical grade and procured from<br />

Ranbaxy Fine Chemicals Ltd., Mumbai,<br />

India. The melting points were determined<br />

in a Toshniwal melting point apparatus<br />

and were uncorrected. The IR spectra of<br />

the compounds were recorded using KBr<br />

pellet method on Rx-1 Perkin-Elmer FTIR.<br />

1 13 HNMR and CNMR spectra were run on<br />

Bruker Avance II 400 spectrophotometer<br />

using CDCl as a solvent. Mass spectra<br />

3<br />

(FAB-MS) were obtained on a JEOL SX<br />

102/DA-6000 mass spectrometer.<br />

Plant Material<br />

The roots of C. spinarum were collected from<br />

Sirsi, Uttara Kannada District, Karnataka,<br />

Research Ar cle<br />

Received Date : 28-11-2011<br />

Revised Date : 03-02-2012<br />

Accepted Date : 08-02-2012<br />

DOI: 10.5530/rjps.2012.1.7<br />

Address for<br />

correspondence<br />

Karunakar Hegde<br />

Department of Pharmacology<br />

H.K.E. Society’s M T R Institute<br />

Srinivas College of Pharmacy<br />

Valachil, Post- Parangepete<br />

Mangalore- 574 143<br />

Karnataka, India<br />

E-mail: khegde_sh2003@<br />

yahoo.co.in<br />

Phone: +91-824-2274722<br />

Fax: +91-824-2274725<br />

www.rjps.in<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 55


Karunakar Hegde et al.: Phytochemical Investigation of Root Extract of the Plant Carissa spinarum<br />

India during May 2007. It was authenticated by<br />

Dr. Gopalakrishna Bhat, Department of Botany, Poorna<br />

Prajna College, Udupi, Karnataka, India. A voucher<br />

specimen no. 105b is deposited in the herbarium of our<br />

institute.<br />

Extraction and Isolation<br />

The shade dried roots of the plant (1000 g) were soaked<br />

in 1.5 L of 95% ethyl alcohol and extracted in the<br />

cold for 4 days with occasional shaking. After 4 days<br />

the ethanol layer was decanted off. The process was<br />

repeated for 4 times. The solvent from the total extract<br />

was fi ltered, the concentrate was evaporated to dryness<br />

under reduced pressure and low temperature (40oC) on a rotary evaporator to give the ethanolic extract<br />

(13% w/w yield), which was stored at 4oC until use.<br />

The ethanol extract (125 g) was suspended in distilled<br />

water and extracted with petroleum ether (60–80oC, 8 ×<br />

500 ml). All the fractions were then washed with distilled<br />

water (30 ml), dried over anhydrous sodium sulphate<br />

and freed of solvent by distillation to give petroleum<br />

ether (60–80ºC) soluble fraction (32 g). Petroleum ether<br />

fraction (25 g) was saponifi ed by refl uxing for 6 h in<br />

500 ml 5% methanolic KOH and allowed to stand at<br />

room temperature for 20 h. The unsaponifi able portion<br />

was extracted with diethyl ether. All the ethereal fractions<br />

Figure 1: Structure of isolated compounds.<br />

were combined and washed with distilled water. The<br />

solvent was evaporated and dried over anhydrous sodium<br />

sulphate to afford a yellowish residue (9 g). 13<br />

The residue (8 g) was dissolved in chloroform (10 ml)<br />

and adsorbed on to silica gel (60–120 mesh, 20 g). After<br />

evaporation of the solvent, it was subjected to column<br />

chromatography over silica gel (150 g) prepared in<br />

n-hexane. The elutions were carried out with n-hexane:<br />

ethyl acetate graded mixture (95:5) afforded compound<br />

I (65 mg) and n-hexane: ethyl acetate graded mixture<br />

(90:10) resulted into compound II (93 mg) and III<br />

(125 mg). Further, the elutions carried out with n-hexane:<br />

ethyl acetate graded mixtures (85:15) resulted into<br />

compound IV (95 mg), V (90 mg) and VI (80 mg)<br />

respectively. The individual compounds were purifi ed<br />

by preparative TLC on silica gel G and further purifi ed<br />

by recrystallization with n-hexane.<br />

RESULTS<br />

Compound I (stigmasterol)<br />

Pearl white crystals (Figure 1); R 0.63 in n-hexane:<br />

f<br />

ethyl acetate (95:05); m.p. 167–170oC; IR v (KBr):<br />

max<br />

3422, 2951, 2851, 1669, 1619, 1465, 1072 cm –1 ; 1HNMR (CDCl , 400 MHz): δ 0.79, 0.78, 0.82, 0.91, 0.94, 1.08<br />

3<br />

(m, 18H), δ 1.13 to δ 2.53 (m, 18H, 9x CH and 8H),<br />

2<br />

56 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Karunakar Hegde et al.: Phytochemical Investigation of Root Extract of the Plant Carissa spinarum<br />

3.54 (1H, dd, J= 9.1, 6.0 Hz, H-α3), 5.33 ( s, 1H, H-3),<br />

5.11 and 5.15 (2H, br); 13 CNMR (CDCl 3, 400 MHz): δ<br />

33.79 (C-1), 78.98 (C-3), δ 145.83 (C-5), δ 121.81 (C-6), δ<br />

32.50 (C-7), δ 35.35 (C-8), δ 45.99 (C-9), δ 30.02 (C-14),<br />

δ 56.93 (C-17), δ 19.91 (C-19), δ 138.39 (C-22), δ129.35<br />

(C-23), δ 30.35 (C-24), δ 18.89 (C-28); FAB-MS m/z<br />

(rel.int): 412 [M + ] (C 29 H 48 O) (100), 397 (20), 369 (10), 351<br />

(70), 329 (65), 300 (40), 299 (23), 273 (31), 255 (28).<br />

Compound II (ursolic acid)<br />

Pale yellow powder (Figure 1); R 0.51 in n-hexane: ethyl<br />

f<br />

acetate (90:10); m.p. 271–274oC; IR v (KBr): 3455, 2924,<br />

max<br />

2853, 1638, 1460, 1382, 1035 cm –1 ; 1HNMR (CDCl , 400 3<br />

MHz): δ 1.00 to 1.91 (m, 22H), 3.20 (1H, dd, J= 10, 5.7<br />

Hz, H-3), 2.17 (1H, d, J= 4.52 Hz, H-18), 2.28 (m, 1H,<br />

OH), 5.34 (s, 1H, H-12); 13CNMR (CDCl 400 MHz):<br />

3,<br />

δ 37.30 (C-1), 78.49 (C-3), 39.98 (C-4), 41.18 (C-5),<br />

51.87 (C-9), 125.25 (C-12), 137.48 (C-13), 55.39 (C-17),<br />

38.95 (C-18), 32.54 (C-19), 18.45 (C-25), 16.95 (C-26),<br />

23.69 (C-27), 177.48 (C-28), 20.12 (C-29); FAB-MS m/z<br />

(rel.int): 456 [M + ] (C H O ) (71), 428 (50), 289 (49),<br />

30 48 3<br />

277 (35), 248 (100), 149 (27), 85 (11), 83 (44).<br />

Compound III (lupeol)<br />

Pearl white crystals (Figure 1); R 0.56 in n-hexane: ethyl<br />

f<br />

acetate (90:10); m.p. 213–215oC; IR v (KBr): 3435,<br />

max<br />

2928, 2850, 1648, 1455, 1373, 1038, 880 cm –1 ; 1HNMR (CDCl , 400 MHz): δ 0.76 to 1.02 (m, 18H, Me-28,<br />

3<br />

Me-23, Me-24, Me-25, Me-26, Me-27), 1.66 (3H, d,<br />

J= 0.7 Hz, Me-30), 3.21 (1H, dd, J= 9.5, 6.2 Hz, H-α3),<br />

4.57 ( s, 1H, H-3); 13CNMR (CDCl 400 MHz): δ 38.55<br />

3,<br />

(C-1), 78.49 (C-3), 38.98 (C-4), 54.34 (C-5), 18.44 (C-6),<br />

39.68 (C-8), 49.87 (C-9), 42.28 (C-14), 42.98 (C-17),<br />

48.24 (C-18), 47.42 (C-19), 150.48 (C-20), 28.25 (C-21),<br />

16.51 (C-25), 15.96 (C-26), 109.29 (C-29); FAB-MS m/z<br />

(rel.int): 426 [M + ] (C H O) (50), 409 (10), 395 (13), 318<br />

30 50<br />

(9), 218 (74), 203 (65), 187 (46), 175 (30), 161 (39), 147<br />

(64), 125 (71), 121 (70), 93 (100).<br />

Compound IV (campesterol)<br />

Pearl white crystals (Figure 1); R 0.79 in n-hexane:<br />

f<br />

ethyl acetate (85:15); m.p. 157–159oC; IR v (KBr):<br />

max<br />

3425, 2935, 2843, 1642, 1474, 1032, 601 cm –1 ; 1HNMR (CDCl , 400 MHz): δ 0.76, to 1.08 (m, 18H, Me-18,<br />

3<br />

Me-21, Me-26, Me-27, Me-24’, Me-19), δ 1.12 to δ 1.26<br />

(m, 16H, 8xCH ), δ 1.39 to δ 1.86 (m, 9H), 3.22 (m, 1H,<br />

2<br />

H-3a), 5.36 (1H, t, J= 3.4 Hz, H-12); 13CNMR (CDCl3, 400 MHz): δ 32.96 (C-1), δ 31.92 (C-2), δ 72.98 (C-3),<br />

δ 150.96 (C-5), δ 124.43 (C-6), δ 50.46 (C-9), δ 40.85<br />

(C-12), δ 43.01 (C-13), δ 29.98 (C-14), δ 56.32 (C-17),<br />

δ 12.66 (C-18), δ 19.80 (C-19), δ 19.39 (C -21), δ 33.86<br />

C-23), δ 31.18 (C-26), δ 21.09 (C-24’); FAB-MS m/z<br />

(rel.int): 400 [M + ] (C H O) (70), 394 (21), 351 (35), 300<br />

28 48<br />

(12), 271 (65), 255 (54), 213 (28), 159 (23), 145 (9), 133<br />

(15), 105 (34), 83 (16), 55 (100 %).<br />

Compound V (17-hydroxy-11-oxo-nor-β-amyrone)<br />

Semisolid compound (Figure 1); R f 0.59 in n-hexane:<br />

ethyl acetate (85:15); m.p. 85–87 o C; IR v max (KBr): 3445,<br />

2931, 1789, 1649, 1454, 1376, 1082, 882 cm –1 ; 1 HNMR<br />

(CDCl 3 , 400 MHz): δ 0.79 (s, 3H, 1xCH 3 , H-27), δ 0.87<br />

(s, 6H, 2xCH 3 , H-25 & H-26), δ 0.91 (s, 6H, 2xCH 3 , H-23<br />

& H-24), δ 0.97 (s, 6H, 2xCH 3 , H-28 & H-29), δ 1.13 to<br />

1.61 (m, 18H, 9xCH 2 ), δ 1.68 to δ 2.31 (s, 3H, 3xCH), δ<br />

3.61 (s, 1H, OH), δ 5.18; 13 CNMR (CDCl 3, 400 MHz): δ<br />

38.94 (C-1), δ 206.82 (C-3), δ 55.34 (C-5), δ 40.02 (C-8),<br />

δ 47.72 (C-9), δ 198.36 (C-11), δ 124.45 (C-12), δ 139.61<br />

(C-13), δ 75.68 (C-17), δ 59.11 (C-18), δ 39.88 (C-19),<br />

δ 39.39 (C-20), δ 41.87 (C-22), δ 28.10 (C-28), δ 15.67<br />

(C-29); FAB-MS m/z (rel.int): 440 [M+H] + (C 29 H 44 O 3 )<br />

(52), 425 (19), 409 (100), 391 (78), 353 (26), 325 (45).<br />

Compound VI (urs-12-ene-3β, 22β-diol-<br />

17-carboxylic acid)<br />

White crystalline powder (Figure 1); R 0.54 in n-hexane:<br />

f<br />

ethyl acetate (85:15); m.p. 211–214oC; IR v (KBr):<br />

max<br />

3455, 2948, 2850, 1642, 1462, 1374, 1055 cm –1 ; 1HNMR (CDCl , 400 MHz): δ 1.00 to 1.91 (m, 23H), δ 2.36 (1H,<br />

3<br />

d, J=7.3 Hz, H-18), δ 3.38 (dd, 1H, OH), δ 3.61 (dd,<br />

1H, OH), δ 5.28 (s, 1H); 13CNMR (CDCl 400 MHz): δ<br />

3,<br />

38.30 (C-1), δ 78.98 (C-3), δ 54.34 (C-5), δ 33.02 (C-7),<br />

δ 39.38 (C-8), δ 46.72 (C-9), δ 123.25 (C-12), δ 139.35<br />

(C-13), δ 41.37 (C-14), δ 48.35 (C-17), δ 38.89 (C-19),<br />

δ 70.68 (C-22), δ 14.99 (C-23), δ 179.33 (C-28), δ 16.61<br />

(C-30); FAB-MS m/z (rel.int): 472 [M] + (C H O ) (100),<br />

30 48 4<br />

456 (45), 428 (12), 289 (30), 277 (35), 241 (70), 234 (25),<br />

149 (9), 85 (42), 83 (20).<br />

DISCUSSION<br />

Compound I was obtained as pearl white crystals<br />

and gave characteristic color reaction for sterol. The<br />

FAB-MS spectrum showed a molecular ion peak at m/z<br />

412 corresponding to molecular formula C H O. The<br />

29 48<br />

IR spectrum exhibited strong absorptions at 3422 cm –1<br />

(hydroxyl group), 2951 and 1669 cm –1 . The 1HNMR spectrum exhibited six tertiary methyl groups at δ 0.79,<br />

0.78, 0.82, 0.91, 0.94, and 1.08, one vinylic proton at δ 5.33<br />

and two olefi nic protons at δ 5.11 and 5.15 respectively.<br />

In 13CNMR spectrum the most down fi eld signals at δ<br />

145.83 was accommodated for sp2 (olefi nic) carbon at<br />

C-5 and the next downfi eld signal at δ 138.39 ppm and<br />

δ 129.35 ppm to C-22 and C-23. The downfi eld signal at<br />

δ 121.81 is to C-6. The oxygenated carbon at C-3 gave<br />

a downfi eld signal at δ 78.98 ppm. The next downfi eld<br />

signal at δ 56.93 ppm was accommodated for C-17.<br />

Other carbon atoms of the steroidal skeleton except<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 57


Karunakar Hegde et al.: Phytochemical Investigation of Root Extract of the Plant Carissa spinarum<br />

that in the side chain appeared in the range δ 45.99 to<br />

δ 30.02 ppm. The angular methyl groups and the side<br />

chain methyl carbons gave signals in the region δ 19.91<br />

to δ 18.89 ppm. 13 CNMR spectral data matched exactly<br />

with that of Stigmasterol 14 . Its identity as stigmasterol<br />

was confi rmed by m.p., IR, 1 HNMR, 13 CNMR and<br />

mass spectral data and by co-chromatography with an<br />

authentic sample.<br />

Compound II was obtained as pale yellow powder and<br />

it gave characteristic color reaction for triterpenoids..<br />

The FAB-MS showed a molecular ion peak at m/z 456<br />

corresponding to a molecular formula C 30 H 48 O 3 . It was<br />

unambiguously identifi ed as ursolic acid on the basis<br />

of its m.p., IR, 1 HNMR, 13 CNMR and mass spectral<br />

data 15 .<br />

Compound III was obtained as pearl white crystals and<br />

it gave characteristic color reaction for triterpenoids.<br />

The FAB-MS spectrum showed a molecular ion peak at<br />

m/z 426 corresponding to molecular formula C 30 H 50 O.<br />

The IR spectrum exhibited strong absorptions at<br />

3435 cm –1 (hydroxyl group) and 2928, 1648, 880 cm –1<br />

(exomethylene group). The 1 HNMR spectrum exhibited<br />

six tertiary methyl groups at δ 0.76, 0.78, 0.82, 0.91,<br />

0.94 and 1.02, a methane group at δ 1.66, a secondary<br />

carbinol group at δ 3.21 and an exomethylene group at<br />

δ 4.56 and 4.68 implies a typical pentacyclic triterpenoid<br />

of the lupeol. Based on the m.p., IR, 1 HNMR, 13 CNMR<br />

and mass spectral data the compound was identifi ed as<br />

lupeol. 16,17<br />

Compound IV obtained as pearl white crystals and it<br />

gave characteristic color reaction for sterol. The peak at<br />

1642.47 cm –1 in IR spectra indicated C=C streching, while<br />

the peaks at 1474.27 cm –1 and 1032.65 cm –1 indicated the<br />

C-H deformation in gem dimethyl and C-O streching of<br />

secondary alcohol respectively. The peak at 3425.25 cm –1<br />

indicated the presence of OH group. The proton NMR<br />

signal at δ 2.17 indicated OH at C–3 position. The signal<br />

at δ 5.36 gave singlet which indicated one vinylic proton<br />

at C-6 position, while the signal at δ 5.12 gave doublet<br />

which indicated two allylic protons at C–7 position. The<br />

13 CNMR spectrum revealed 28 signals which were duly<br />

assigned as CH 3 , CH 2 , CH and -C- group represent in the<br />

compound. The FAB-MS spectrum showed a molecular<br />

ion peak at m/z 400 corresponding to molecular formula<br />

C 28 H 48 O. The mass fragmentation was typical to that of<br />

campesterol. Based on the m.p., IR, 1 HNMR, 13 CNMR<br />

and mass spectral data the compound was identifi ed as<br />

campesterol.<br />

Compound V obtained as semisolid mass and gave<br />

characteristic color reaction for triterpenoids. The peak at<br />

1789.68 cm –1 in IR spectra indicated C=O streching. The<br />

peak at 882.12 cm –1 indicated =C-H out plane bending.<br />

The peak at 1649.64 cm –1 indicated C=C streching and<br />

peak at 1376.69 cm –1 indicated the C-H deformation<br />

in gem dimethyl. The peak at 3445.42 cm –1 indicated<br />

the presence of OH group. The proton NMR signals<br />

at δ 5.18 indicates vinylic protons and another singlet<br />

at δ 3.61 indicated OH at C-17 position. The 13 CNMR<br />

spectrum revealed 29 signals which were duly assigned<br />

as CH 3 , CH 2 , CH, -C- and -C=C- group represent in the<br />

compound. The FAB-MS spectrum showed a molecular<br />

ion peak at m/z 400 corresponding to molecular formula<br />

C 29 H 44 O 3 . The mass fragmentation was typical to that<br />

of β-amyrone skeleton. From the above evidences the<br />

compound was identifi ed as 17-Hydroxy-11-oxo-norβ-amyrone<br />

18 .<br />

Compound VI obtained as white crystals and gave a<br />

characteristic color reaction for triterpenoids. The FAB-<br />

MASS spectrum displayed the characteristic retro-Diels-<br />

Alder fragment peak at m/z 234 indicating a C-12 /<br />

C-13 double bond which suggested an ursane structure 19<br />

substituted by two hydroxyl groups (-OH), one located<br />

at A/B rings and one on D/E rings. These results<br />

are in good agreement with the IR spectrum, which<br />

showed absorption bands at 3455.84 cm –1 assigned to<br />

the hydroxyl groups and 1642.89 cm –1 corresponds to<br />

carbonyl group of the carboxylic acid. The 1 HNMR<br />

spectrum displayed seven methyl signals. In addition,<br />

the presence of the olefi nic triplet at δ 5.28 and two<br />

hydroxyl bearing methine protons exhibiting two double<br />

doublets at δ 3.38 and δ 3.61. On the basis of the<br />

1 HNMR spectrum of the compound, a β-confi guration<br />

of the two hydroxyl groups was assigned according to<br />

the fact that their respective geminal protons appeared<br />

as dd thus confi rming axial orientation. 20 Further the<br />

13 CNMR spectrum revealed 30 signals which were duly<br />

assigned as CH 3 , CH 2 , CH and -C- group represent in the<br />

compound. Therefore the structure of compound VI is<br />

identifi ed as Urs-12-ene-3β, 22β-diol-17-carboxylic acid,<br />

whose mass value obtained as 472 by FAB-MASS which<br />

corresponds to the C 30 H 48 O 4 ; the molecular formula of<br />

the compound.<br />

CONCLUSION<br />

From the spectral data, the compounds I-VI was analyzed<br />

as stigmasterol, ursolic acid, lupeol, campesterol,<br />

17-hydroxy-11-oxo-nor-β-amyrone and urs-12-ene-3β,<br />

22β-diol-17-carboxylic acid respectively. However, the<br />

compounds stigmasterol, campesterol, 17-hydroxy-11oxo-nor-β-amyrone<br />

and urs-12-ene-3β, 22β-diol-17carboxylic<br />

acid were reported for the fi rst time from the<br />

root of this plant.<br />

REFERENCES<br />

1. Kirtikar KR, Basu BD. Indian Medicinal Plants, Vol. II, Lalit Mohan Basu,<br />

Allahabad, 2003,1548–9.<br />

2. Chopra RN, Nayar SL, Chopra IC. Glossary of Indian Medicinal Plants,<br />

(CSIR, New Delhi), 1956;52.<br />

58 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Karunakar Hegde et al.: Phytochemical Investigation of Root Extract of the Plant Carissa spinarum<br />

3. Vohra MM, De NN. Comparative cardiotonic activity of Carissa carandas<br />

and Carissa spinarum. Indian J Med Res 1963;51:937–40.<br />

4. Karunakar Hegde, D Satyanarayana, Arun B Joshi. Evaluation of<br />

anticonvulsant activity of Carissa spinarum root extract. RGUHS<br />

J Pharm Sci 2011;1:64–8.<br />

5. Karunakar Hegde, Arun B Joshi. Hepatoprotective and antioxidant effect<br />

of Carissa spinarum root extract against CCl 4 and paracetamol induced<br />

hepatic damage in rats. Bangladesh J Pharmacol 2010;5:73–6.<br />

6. Karunakar Hegde, Cijo Issac, Arun B Joshi. Antiarthritic activity of<br />

Carissa spinarum root extract in Freund’s adjuvant induced polyarthritis<br />

in rats. Pharmacologyonline 2010;2:713–8.<br />

7. Mathuram V, Brahmadhayalaselvam A, Hussain AJ, Rao RB, Patra<br />

A. Chemical constituents of Carissa spinarum and their antibacterial<br />

activity. J Indian Chem Soc 1998;75:262–4.<br />

8. Rao RJ, Kumar US, Reddy SV, Tiwari AK, Rao JM. Antioxidants and a<br />

new germacrane sesquiterpene from Carissa spinarum. Nat Prod Res<br />

2005;19:763–9.<br />

9. Bhakuni DS, Dhar ML, Dhar MM, Dhawan BN, Gupta B, Srimal RC.<br />

Screening of Indian plants for biological activity: Part III. Indian J Exptl<br />

Biol 1971;9:91–102.<br />

10. Rastogi RC, Kulshreshtha DK, Rastogi RP. Cardioactive constituents<br />

from Carissa spinarum Linn. Indian J Chem 1969;7:1102–4.<br />

11. Raina MK, Bhatnaga JK, Atal CK. Isolation of caffeic acid from the roots<br />

of Carissa spinarum Linn. The Indian J Pharm 1971;33:76–7.<br />

12. Mathuram V, Brahmadhayalaselvam A, Hussain AJ, Rao RB, Patra<br />

A. Chemical constituents of Carissa spinarum and their antibacterial<br />

activity. J Indian Chem Soc 1998;75:262–4.<br />

13. Pearlman WH, Emily C. The isolation of C 21 steroids from human<br />

placenta. J Biol Chem 1951;8:807–14.<br />

14. Sright JLC, McInnes AG, Shimizu S, et al. Identifi cation of C-24<br />

alkyl epimers of marine sterols by 13C nuclear magnetic resonance<br />

spectroscopy. Can J Chem 1978;56:1898–1903.<br />

15. Saeidnia S, Gohari AR, Uchiyama N, Ito M, Honda G, Kiuchi F. Tow<br />

new monoterpene glycosides and trypanocidal terpenoids from<br />

Dracocephalum kotschyi. Chem Pharm Bull 2004;52:1249–50.<br />

16. Gunasekera SP, Cordell GA, Farnsworth NR. Constituents of<br />

Pithecellobium multifl orum. J Nat Prod 1982;45:651–4.<br />

17. Garcia B, Alberto Macro J, Seoane E, Tortajada A. Triterpinoids, waxes<br />

and tricin in Phoenix canariensis. J Nat Prod 1981;44:111–3.<br />

18. Atlas of mass spectra of triterpenoid compound in varnish. Available<br />

from: URL: http://www.amolf.nl/publications/theses/doelen/Atlas.pdf<br />

19. Budzikiewicz H, Wilson JM, Djerassi C. Mass spectrometry in structural<br />

and stereochemical problems. J Am Chem Soc 1963;85:3688–99.<br />

20. Machocho AK, Kiprono PC, Grinberg S, Bittner S. Pentacyclic<br />

triterpenoids from Emblica schimperi. Phytochem 2003;62:573–7.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 59


Synthesis and antimicrobial activity of<br />

4-hydroxy-1-methyl/phenyl-3- (substituted<br />

anilinoacetyl) quinolin-2(1H)-one<br />

Girish Bolakatti 1 , Manjunatha S. Katagi* 1 , S.N. Mamledesai 2 , Sujatha. M.L. 3,<br />

Prakash Dabadi 1 , Narayana Miskin. 1<br />

1 Bapuji Pharmacy College, Davangere 577 004, Karnataka, India<br />

2 P.E.S’s College of Pharmacy education and Research, Ponda 403 401, Goa, India<br />

3 Bapuji Institute of Engineering and Technology, Davangere 577 004, Karnataka, India<br />

ABSTRACT<br />

A series of new 2-quinolone derivatives were synthesized, purifi ed and characterized on the basis of IR, 1H NMR, 13C NMR and Mass spectral studies. Same compounds were evaluated for antimicrobial activity against<br />

Staphylococcus aureus, Bacillus substilis, Escherichia coli, Pseudomonas aeruginosa, Candida albicans and<br />

Asparagillus niger. Among eighteen synthesized novel compounds, in which fi ve compounds (3a, 3b, 3d, 4a, 4c)<br />

shows promising antibacterial activity as compared to Ciprofl oxacin (100 μg/ml), however most of them (3a, 3b,<br />

3d, 3e, 4a, 4b, 4d, 4e) showed potent antifungal activity as compared to Fluconazole (100 μg/ml).<br />

Key words: Mass spectral, 2-quinolone, antibacterial and antifungal<br />

INTRODUCTION<br />

Although considerable advances have been<br />

achieved over recent decades in the research<br />

and development of new structural prototypes<br />

as effective antimicrobials, current antimicrobial<br />

chemotherapy still suffers from<br />

two major limitations. The fi rst is the lack<br />

of selectivity of conventional antimicrobial<br />

agents, which in turn brings about unwanted<br />

side effects. The second is acquisitions by<br />

the microorganism of multi drug resistance.<br />

The design of new agents, active against<br />

resistant organism is of critical importance.<br />

In the fi eld of quinolone antimicrobial<br />

agents, the new generation of quinolone<br />

has achieved signifi cant improvements in<br />

terms of potency, spectrum and pharmacokinetic<br />

properties. But these agents faced<br />

a rapid increase of resistance from grampositive<br />

organisms. Therefore, enhancing<br />

the potency of quinolone especially against<br />

gram-positive organism has become most<br />

urgent. 1<br />

The resistance to antimicrobial drugs is<br />

wide spread, the development of new<br />

antimicrobial agents and understanding<br />

their mechanisms of action are becoming<br />

vital nowadays. Nitrogen containing heterocyclic<br />

compounds is an indispensable<br />

structural unit for both the chemist and<br />

the biochemist. Among the antimicrobial<br />

agents discovered in recent years the various<br />

2-quinolone as potent and selective antimicrobial<br />

agent has stimulated remarkable<br />

interest in the synthesis of 2-quinolones<br />

bearing heterocycles. 2 Numerous biological<br />

activities of 2-quinolone have been<br />

described; antimicrobial, 3–5 antioxidant and<br />

anti-infl ammatory, 6 antitumor, 7 Fornesyl<br />

transferase inhibitor 8 , antiangiogenic 9 and<br />

anti-tuberculosis. 10–13<br />

EXPERIMENTAL PROCEDURE<br />

All the chemicals and solvents were<br />

supplied by Merck, S.D Fine-Chem.<br />

Research Ar cle<br />

Received Date : 19-11-2011<br />

Revised Date : 02-03-2012<br />

Accepted Date : 05-03-2012<br />

DOI: 10.5530/rjps.2012.1.8<br />

Address for<br />

correspondence<br />

Manjunatha S. Katagi<br />

Bapuji Pharmacy College<br />

S.S. Layout<br />

Davangere-577 004<br />

Karnataka, India<br />

Phone: 08192-221459<br />

Mobile: +919886499160<br />

Fax: 08192-222561<br />

Email: manju_mpharm@<br />

rediffmail.com<br />

www.rjps.in<br />

60 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Girish Bolakatti et al.: Synthesis and antimicrobial activity of 4-Hydroxy-1-methyl/phenyl-3- (Substituted anilinoacetyl) quinolin-2(1H)-one<br />

Limited, Mumbai. All the solvents were distilled before<br />

use and chemicals were purifi ed by either distillation or<br />

recrystallisation before use. The melting points were<br />

taken on the Veego (VMP–MP) melting point apparatus<br />

and are uncorrected. The IR spectra of the compounds<br />

were recorded using KBr on Jasco FTIR spectrometer<br />

(model-4100). The 1 H NMR and CHN elemental<br />

analysis of the synthesized compounds was recorded on<br />

Bruker avance II 400 NMR spectrometer (with TMS as<br />

internal references) and Perkin Elmer 2400 respectively<br />

at Sophisticated Analytical and Instrumentation Facility<br />

(SAIF), Punjab University (Chandigarh). Mass spectra<br />

were recorded on Shimadzu LC MS-2010A at Quest<br />

Research and Training Institute (Pvt) Ltd, Bangalore.<br />

4-Hydroxy-6-methyl/phenyl-2H-pyrano [3,2-c] quinoline-<br />

2,5(6H)-dione, were synthesized according to literature<br />

procedure and subjected to hydrolysis to yield 1a and<br />

1b, 14 further 1a and 1b subjected to bromination to yield<br />

3-Bromoacetyl-4-hydroxy-1-methyl/phenyl quinolin-<br />

2(1H)-one 2a and 2b. The synthesis of title compounds<br />

3a-h and 4a-h were accomplished by condensing with<br />

different substituted aniline. The compounds thus<br />

obtained were characterized by IR, 1 HNMR and mass<br />

spectral data. The physicochemical properties of titled<br />

compounds were given in Table 1 and Table 2.<br />

General procedure for synthesis of<br />

3-(Bromoacetyl)-4-hydroxy-1-methyl/<br />

phenylquinolin-2(1H )-one 2a, 2b<br />

A solution of 3-Acetyl-4-hydroxy-1-methyl/phenyl<br />

quinolin-2(1H )-one (0.065 mol) in glacial acetic acid<br />

(50 ml) was heated (80–100°)( follow the instruction<br />

provided on page no 5 of instruction to authors) with<br />

constant stirring, then bromine (0.065 mol) in glacial<br />

acetic acid (20 ml) was added drop wise over a period<br />

of 1 h and heated the solution until slight change in the<br />

color of the solution was seen. The reaction mixture was<br />

then cooled to room temperature. The product obtained<br />

as yellow crystals and recrystallized from ethanol.<br />

3-(Bromoacetyl)-4-hydroxy-1-methylquinolin-<br />

2(1H )-one 2a<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 10.26 g (73%), mp 174–176<br />

°C, IR (KBr, v, cm –1 ): 756.14 cm –1 and 1190.18 (CH 2 -Br),<br />

1615.47 cm –1 (– C = O amide), 3046.24cm –1 (aromatic<br />

–C–H stre). 1 H NMR (400MHz, CDCl 3 ) δ (ppm): δ<br />

3.78 (s, 3H, N–CH 3 ), 4.98 (s, 2H, CH 2 –Br), 7.72–8.35<br />

(m, 4H, Ar–H), 15.82 (s, 1H, OH).<br />

3-(Bromoacetyl)-4-hydroxy-1-phenylquinolin-<br />

2(1H )-one 2b<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 12.24 g (67.43%), mp<br />

192–194°, IR (KBr, v, cm –1 ): 786.11 cm –1 and 1210.06<br />

(CH 2 -Br), 1620.31 cm –1 (– C = O amide), 3058.11cm –1<br />

(aromatic –C – H stre). 1 H NMR (400MHz, CDCl 3 ) δ<br />

(ppm): δ 5.23 (s, 2H, CH 2 -Br), 7.65–8.85 (m, 9H, Ar–H),<br />

15.91 (s, 1H, OH).<br />

General procedure for synthesis of 4-Hydroxy-<br />

1-methyl/phenyl-3-(Substituted anilinoacetyl)<br />

quinolin-2(1H )-one 3a-h and 4a-h<br />

A solution of 3-(Bromoacetyl)-4-hydroxy-1-methyl/<br />

phenylquinolin-2(1H)-one 2a, 2b (0.004 mol) in acetone<br />

Table 1: Characterization data of 4-Hydroxy-1-methyl -3-(Substituted anilinoacetyl)-quinolin-2(1H)-one 3a-h<br />

Analysis( % )Found (Calcd) in % Yield<br />

Comp R Mol formula M.P. °C * Rf C H N<br />

3a -2,5-Dichloro C H Cl N O 18 14 2 2 3 210–212 0.56 57.31 (57.16) 3.74 (3.61) 7.43 (7.37)<br />

3b -2,4-Dichloro C H Cl N O 18 14 2 2 3 206–208 0.53 57.31 (57.19) 3.74 (3.64) 7.43 (7.39)<br />

3c -p-Nitro C H N O 18 15 3 5 198–200 0.47 61.19 (61.08) 4.28 (4.21) 11.89 (11.79)<br />

3d -p-Fluro C H FN O 18 15 2 3 204–206 0.41 66.25 (66.18) 4.63 (4.51) 8.58 (8.49)<br />

3e p-Chloro C H ClN O 18 15 2 3 211–213 0.42 63.07 (63.01) 4.41 (4.35) 8.17 (8.11)<br />

3f -3-Chloro-4-fl uro C H ClFN O 18 14 2 3 220–222 0.77 59.93 (59.85) 3.91 (3.86) 7.76 (7.71)<br />

3g -2,4-Dimethyl C H N O 20 20 2 3 225–227 0.51 71.41 (71.37) 5.99 (5.91) 8.33 (8.24)<br />

3h -3,4-Dimethyl C H N O 20 20 2 3 222–224 0.53 71.41 (71.35) 5.99 (5.92) 8.33 (8.25)<br />

*TLC Solvent system: Chloroform: Methanol: Strong ammonia-(10:5:3) .<br />

OH O<br />

N<br />

CH 3<br />

3a-h<br />

O<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 61<br />

NH<br />

R


Girish Bolakatti et al.: Synthesis and antimicrobial activity of 4-Hydroxy-1-methyl/phenyl-3- (Substituted anilinoacetyl) quinolin-2(1H)-one<br />

Table 2: Characterization data of 4-Hydroxy-1-phenyl -3-(Substituted anilinoacetyl) quinolin-2(1H)-one 4a-h<br />

(50 ml) was cooled at 0º, primary amine (0.004 mol)<br />

dissolved in acetone (25 ml) was added with stirring<br />

at 0–5° followed by drop wise addition of sodium<br />

hydroxide (0.004 mol) in water (25 ml). Contents were<br />

stirred for 3 hour and poured in to ice water acidifi ed<br />

with dil. HCl, fi ltered, washed, dried and purifi ed by<br />

crystallization from a ethanol + water (9:1).<br />

3-{2-[(2,5-dichlorophenyl)amino]acetyl}-4hydroxy-1-methylquinolin-2(1H)-one<br />

3a<br />

This was prepared and purified as per the above<br />

mentioned procedure: yield 0.75 g (49.73%); IR<br />

(KBr, v, cm –1 ): 1617.61 cm –1 (– C = O amide), 3156.20<br />

cm –1 (– CH stre), 3266.33 cm –1 (– NH), 3363.09 cm –1<br />

(– OH); 1 H NMR (400MHz, DMSO-d 6 ) δ (ppm):<br />

2.85 (s, 3H, N- CH 3 ), 3.87 (s, 1H, NH), 4.81 (s, 2H,<br />

COCH 2 ), 6.45–7.5 (m, 7H, Ar–H), 15.24 (s, 1H,<br />

OH); 13 C NMR (400MHz, DMSO-d 6 ) δ (ppm): 31.5<br />

(N-CH 3 ), 59.8 (CH 2 ), 108.01 115.12, 115.65, 119.32,<br />

121.23, 121.93, 124.99, 126.11, 128.84, 132.36,<br />

134.24, 135.85, 145.40 (Aromatic carbons), 164.53<br />

(exocyclic ketone), 166.32 (Hydroxyl carbon), 196.85<br />

(Carbonyl carbon); LCMS: C 18 H 14 Cl 2 N 2 O 3 (M + ) m/z<br />

377.29; calcd. 377.22.<br />

Analysis( % )Found (Calcd) in % Yield<br />

Comp R Mol formula M.P. 0C * Rf C H N<br />

4a -2,5-Dichloro C H Cl N O 23 16 2 2 3 240–242 0.58 62.88 (62.81) 3.67 (3.60) 6.38 (6.32)<br />

4b -2,4-Dichloro C H Cl N O 23 16 2 2 3 236–238 0.55 62.88 (62.83) 3.67 (3.62) 6.38 (6.34)<br />

4c -p-Nitro C 23 H 17 N 3 O 5 245–247 0.45 66.50 (66.41) 4.12 (4.01) 10.12 (10.01)<br />

4d -p-Fluro C 23 H 17 FN 2 O 3 251–253 0.43 71.13 (71.02) 4.41 (4.32) 7.21 (7.16)<br />

4e p-Chloro C 23 H 17 ClN 2 O 3 211–213 0.45 68.23 (68.17) 4.23 (4.14) 6.92 (6.87)<br />

4f -3-Chloro-4-fl uro C 23 H 16 ClFN 2 O 3 208–210 0.73 65.33 (65.23) 3.81 (3.74) 6.63 (6.52)<br />

4g -2,4-Dimethyl C H N O 25 22 2 3 246–248 0.47 75.36 (75.23) 5.57 (5.48) 7.03 (6.97)<br />

4h -3,4-Dimethyl C H N O 25 22 2 3 251–253 0.49 75.36 (75.28) 5.57 (5.45) 7.03 (6.98)<br />

* TLC Solvent system: Chloroform: Methanol: Strong ammonia-(10:5:3).<br />

OH O<br />

N<br />

3-{2-[(2,4-dichlorophenyl)amino]acetyl}-4hydroxy-1-methylquinolin-2(1H)-one<br />

3b<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.71 g (47.08%); IR (KBr,<br />

v, cm –1 ): 1620.54 cm –1 (– C = O amide), 3158.51 cm –1<br />

(– CH stre), 3271.31 cm –1 (– NH), 3359.05 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 2.82 (s, 3H,<br />

N–CH 3 ), 3.85 (s, 1H, NH), 4.75 (s, 2H, COCH 2 ), 6.35–<br />

7.58 (m, 7H, Ar–H), 15.04 (s, 1H, OH).<br />

3-[2-(4-Nitrophenylamino)acetyl]-4-hydroxy-<br />

1-methylquinolin-2(1H)-one 3c<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.86 g (60.90%); IR (KBr,<br />

v, cm –1 ): 1623.11 cm –1 (– C = O amide), 3186.22 cm –1<br />

(– CH stre), 3252.38 cm –1 (– NH), 3345.23 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 2.91 (s, 3H, N-<br />

CH 3 ), 4.06 (s, 1H, NH), 4.96 (s, 2H, COCH 2 ), 6.86–7.92<br />

(m, 8H, Ar–H), 14.52 (s, 1H, OH); 13 C NMR (400MHz,<br />

DMSO-d 6 ) δ (ppm): 32.5 (N-CH 3 ), 60.2 (CH 2 ), 107.25,<br />

114.16, 115.21, 121.76, 121.99, 124.65, 126.11, 128.84,<br />

135.05, 136.56, 153.65 (Aromatic carbons), 164.36<br />

(exocyclic ketone), 166.32 (Hydroxyl carbon), 196.12<br />

62 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012<br />

O<br />

NH<br />

4a-h<br />

R


Girish Bolakatti et al.: Synthesis and antimicrobial activity of 4-Hydroxy-1-methyl/phenyl-3- (Substituted anilinoacetyl) quinolin-2(1H)-one<br />

(Carbonyl carbon) ; LCMS: C 18 H 15 N 3 O 5 (M + ) m/z<br />

353.33; calcd. 353.32.<br />

3-[2-(4-Fluorophenylamino)acetyl]-4-hydroxy-<br />

1-methylquinolin-2(1H)-one 3d.<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.56 g (42.94%); IR (KBr,<br />

v, cm –1 ): 1613.16 cm –1 (– C = O amide), 3180.01 cm –1<br />

(– CH stre), 3245.02 cm –1 (– NH), 3305.03 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 2.75 (s, 3H, N-<br />

CH 3 ), 3.89 (s, 1H, NH), 4.53 (s, 2H, COCH 2 ), 6.45–7.53<br />

(m, 8H, Ar–H), 14.96 (s, 1H, OH).<br />

3-[2-(4-Chlorophenylamino)acetyl]-4-hydroxy-<br />

1-methylquinolin-2(1H)-one 3e<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.68 g (49.63%); IR (KBr,<br />

v, cm –1 ): 1613.51 cm –1 (– C = O amide), 3123.26 cm –1<br />

(– CH stre), 3243.36 cm –1 (– NH), 3345.12 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 2.63 (s, 3H, N-<br />

CH 3 ), 3.91 (s, 1H, NH), 4.56 (s, 2H, COCH 2 ), 6.49–7.62<br />

(m, 8H, Ar–H), 14.99 (s, 1H, OH).<br />

3-[2-(3-Chloro-4-fl uorophenylamino)acetyl]-<br />

4-hydroxy-1-methylquinolin-2(1H)-one 3f<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.78 g (54.09%); IR (KBr,<br />

v, cm –1 ): 1613.05 cm –1 (– C = O amide), 3123.12 cm –1<br />

(– CH stre), 3243.56 cm –1 (– NH), 3355.10 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 2.83 (s, 3H, N-<br />

CH 3 ), 3.99 (s, 1H, NH), 4.63 (s, 2H, COCH 2 ), 6.49–7.51<br />

(m, 7H, Ar–H), 14.82 (s, 1H, OH).<br />

3-[2-(2,4-dimethylphenylamino)acetyl]-4-hydroxy-<br />

1-methylquinolin-2(1H)-one 3g<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.81 g (60.26%); IR (KBr, v,<br />

cm –1 ): 1632.05 cm –1 (– C = O amide), 3178.28 cm –1 (– CH<br />

stre), 3288.88 cm –1 (– NH), 3355.51 cm –1 (– OH); 1 H NMR<br />

(400MHz, DMSO-d 6 ) δ (ppm): 2.32 (s, 6H, CH 3 ), 2.42 (s,<br />

3H, N- CH 3 ), 3.75 (s, 1H, NH), 4.85 (s, 2H, COCH 2 ),<br />

6.38–7.53 (m, 7H, Ar–H), 15.23 (s, 1H, OH); 13 C NMR<br />

(400MHz, DMSO-d 6 ) δ (ppm): 21.9, 24.6 (2 CH 3 ), 31.5<br />

(N–CH 3 ), 60.2 (CH 2 ), 107.25, 113.16, 115.21, 121.5,<br />

121.76, 124.99, 126.76, 126.96, 128.84, 131.75, 135.05,<br />

143.65 (Aromatic carbons), 164.10 (exocyclic ketone),<br />

166.82 (Hydroxyl carbon), 196.65 (Carbonyl carbon) ;<br />

LCMS: C 20 H 20 N 2 O 3 (M + ) m/z 336.32; calcd. 336.38.<br />

3-[2-(3,4-dimethylphenylamino)acetyl]-4-hydroxy-<br />

1-methylquinolin-2(1H)-one 3h<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.96 g (71.42%); IR (KBr,<br />

v, cm –1 ): 1632.00 cm –1 (– C = O amide), 3178.28 cm –1<br />

(– CH stre), 3288.88 cm –1 (– NH), 3355.51 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 2.15 (s, 6H,<br />

CH 3 ), 2.42 (s, 3H, N- CH 3 ), 3.75 (s, 1H, NH), 4.85 (s, 2H,<br />

COCH 2 ), 6.38–7.53 (m, 7H, Ar–H), 15.23 (s, 1H, OH).<br />

3-{2-[(2,5-dichlorophenyl)amino]acetyl}-4hydroxy-1-phenylquinolin-2(1H)-one<br />

4a<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.95 g (54.10%); IR (KBr, v,<br />

cm –1 ): 1625.61 cm –1 (– C = O amide), 3166.00 cm –1 (– CH<br />

stre), 3260.83 cm –1 (– NH), 3392.08 cm –1 (– OH); 1 H<br />

NMR (400MHz, DMSO-d 6 ) δ (ppm): 3.89 (s, 1H, NH),<br />

4.76 (s, 2H, COCH 2 ), 6.60–7.64 (m, 12H, Ar–H), 15.20<br />

(s, 1H, OH); 13 C NMR (400MHz, DMSO-d 6 ) δ (ppm):<br />

59.91 (CH 2 ), 107.5, 113.01, 115.22, 118.21, 118.32,<br />

118.93, 119.21, 119.31, 120.23, 127.84, 128.36, 129.7,<br />

131.1, 133.25, 137.2, 140.7, 145.4 (Aromatic carbons),<br />

159.53 (exocyclic ketone), 166.30 (Hydroxyl carbon),<br />

196.85 (Carbonyl carbon) ; LCMS: C 23 H 16 Cl 2 N 2 O 3 (M + )<br />

m/z 439.23; calcd. 439.29.<br />

3-{2-[(2,4-dichlorophenyl)amino]acetyl}-<br />

4-hydroxy-1-phenylquinolin-2(1H)-one 4b<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.92 g (52.39%); IR (KBr,<br />

v, cm –1 ): 1620.54 cm –1 (– C = O amide), 3158.51 cm –1<br />

(– CH stre), 3271.31 cm –1 (– NH), 3359.05 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 3.93 (s, 1H,<br />

NH), 4.81 (s, 2H, COCH 2 ), 6.63–7.69 (m, 12H, Ar–H),<br />

15.05 (s, 1H, OH).<br />

3-[2-(4-Nitrophenylamino)acetyl]-4-hydroxy-<br />

1-phenylquinolin-2(1H)-one 4c<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 1.06 g (63.85%); IR (KBr,<br />

v, cm –1 ): 1628.65 cm –1 (– C = O amide), 3145.06 cm –1<br />

(– CH stre), 3245.38 cm –1 (– NH), 3354.23 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 4.06 (s, 1H,<br />

NH), 4.82 (s, 2H, COCH 2 ), 6.86–7.92 (m, 13H, Ar–H),<br />

14.82 (s, 1H, OH).<br />

3-[2-(4-Fluorophenylamino)acetyl]-4-hydroxy-<br />

1-phenylquinolin-2(1H)-one 4d<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.49 g (31.57%); IR (KBr,<br />

v, cm –1 ): 1619.13 cm –1 (– C = O amide), 3188.05 cm –1<br />

(– CH stre), 3251.02 cm –1 (– NH), 3389.03 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 4.15 (s, 1H,<br />

NH), 4.91 (s, 2H, COCH 2 ), 7.12–8.13 (m, 13H, Ar–H),<br />

14.92 (s, 1H, OH).<br />

3-[2-(4-Chlorophenylamino)acetyl]-4-hydroxy-<br />

1-phenylquinolin-2(1H)-one 4e.<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.56 g (34.61%); IR<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 63


Girish Bolakatti et al.: Synthesis and antimicrobial activity of 4-Hydroxy-1-methyl/phenyl-3- (Substituted anilinoacetyl) quinolin-2(1H)-one<br />

(KBr, v, cm –1 ): 1619.51 cm –1 (– C = O amide), 3187.26<br />

cm –1 (– CH stre), 3256.31 cm –1 (– NH), 3385.18 cm –1<br />

(– OH); 1 H NMR (400MHz, DMSO-d 6 ) δ (ppm): 4.11<br />

(s, 1H, NH), 4.67 (s, 2H, COCH 2 ), 7.05–8.11 (m, 13H,<br />

Ar–H), 14.91 (s, 1H, OH).<br />

3-[2-(3-Chloro-4-fl uorophenylamino)acetyl]-<br />

4-hydroxy-1-phenylquinolin-2(1H)-one 4f<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.88 g (52.07%); IR (KBr,<br />

v, cm –1 ): 1610.05 cm –1 (– C = O amide), 3142.02 cm –1<br />

(– CH stre), 3251.16 cm –1 (– NH), 3420.15 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 3.85 (s, 1H,<br />

NH), 4.68 (s, 2H, COCH 2 ), 6.84–8.11 (m, 12H, Ar–H),<br />

13.92 (s, 1H, OH).<br />

3-[2-(2,4-dimethylphenylamino)acetyl]-<br />

4-hydroxy-1-phenylquinolin-2(1H)-one 4g<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.83 g (52.13%); IR (KBr,<br />

v, cm –1 ): 1653.12 cm –1 (– C = O amide), 3251.06 cm –1<br />

(– CH stre), 3296.80 cm –1 (– NH), 3395.22 cm –1 (– OH).<br />

3-[2-(3,4-dimethylphenylamino)acetyl]-<br />

4-hydroxy-1-phenylquinolin-2(1H)-one 4h<br />

This was prepared and purifi ed as per the above<br />

mentioned procedure: yield 0.96 g (60.30%); IR (KBr,<br />

v, cm –1 ): 1612.00 cm –1 (– C = O amide), 3178.33 cm –1<br />

(– CH stre), 3269.28 cm –1 (– NH), 3358.01 cm –1 (– OH);<br />

1 H NMR (400MHz, DMSO-d6 ) δ (ppm): 2.13 (s, 6H,<br />

CH 3 ), 3.78 (s, 1H, NH), 4.96 (s, 2H, COCH 2 ), 6.98–8.35<br />

(m, 12H, Ar–H), 14.01 (s, 1H, OH).<br />

ANTIMICROBIAL SCREENING15–18 All the synthesized compounds 3a-h and 4a-h have<br />

been screened in vitro for their antibacterial activity<br />

against gram-negative bacteria Escherichia coli (ATCC<br />

10536), Pseudomonas aeruginosa (ATCC 10145) and grampositive<br />

bacteria Staphylococcus aureus (ATCC11632),<br />

Bacillus substilis (ATCC 60511), while antifungal activity<br />

against Candida albicans (ATCC 2501) and Asparagillus<br />

niger (ATCC 1781) at 100 μg/ml concentration by cupplate<br />

agar diffusion method using dimethylsulfoxide<br />

as solvent. After 24 and 48h of incubation at 37° ±1,<br />

the antibacterial and antifungal activity respectively<br />

was determined by measuring the zones of inhibition<br />

in mm. Standard antibacterial ciprofl oxacin and<br />

fungicide fl uconazole were used under similar condition<br />

for comparison. Control test with solvent were<br />

performed for every assay but showed no inhibition<br />

of microbial growth. The observed zone of inhibition<br />

for antibacterial and antifungal activity is presented in<br />

Table 3<br />

Table 3: In vitro antimicrobial activities of the 2-quinolone<br />

derivatives at a concentration of 100 µg/ml (zone of<br />

inhibition in mm)<br />

Zone of inhibition<br />

Compounds Sa Bs Ec Pa Ca An<br />

3a 19 22 29 28 28 33<br />

3b 18 23 28 26 30 34<br />

3c 10 12 13 NA 16 18<br />

3d 20 23 25 27 31 34<br />

3e 09 07 10 08 27 33<br />

3f 07 08 NA 10 15 16<br />

3g 08 10 13 15 17 12<br />

3h 11 13 NA 17 13 19<br />

4a 19 24 31 29 28 31<br />

4b 11 12 11 13 27 30<br />

4c 21 25 32 30 11 16<br />

4d 12 16 19 21 29 28<br />

4e 09 07 10 11 32 34<br />

4f 12 11 13 NA 13 15<br />

4g 09 07 10 11 NA NA<br />

4h 08 NA NA NA 10 NA<br />

Ciprofl oxacin 23 26 34 31 ND ND<br />

Fluconazole ND ND ND ND 33 36<br />

Sa = Staphylococcus aureus, Bs = Bacillus substilis, Ec = Escherichia coli,<br />

Pa = Pseudomonas aeruginosa, Ca = Candida albicans, An = Asparagillus niger, NA = No<br />

activity, ND = Not determined.<br />

RESULTS AND DISCUSSION<br />

4-Hydroxy-6-methyl/phenyl-2H-pyrano[3,2-c]<br />

quinoline-2,5(6H)-dione, were synthesized according to<br />

literature procedure14 and subjected to hydrolysis to yield<br />

1a and 1b, further 1a and 1b subjected to bromination<br />

to yield 3-Bromoacetyl-4-hydroxy-1-methyl/phenyl<br />

quinolin-2(1H)-one 2a and 2b. The synthesis of title<br />

compounds 3a-h and 4a-h were accomplished by<br />

condensing with different substituted aniline.<br />

The structures of newly synthesized compounds were<br />

confi rmed by their spectral data. The characteristic<br />

IR absorption peaks 1617.61 cm –1 was due to amide<br />

carbonyl, 3156.20 cm –1 mainly because aromatic –C – H<br />

stretching, 3266.33 cm –1 may be due to free amine group<br />

of acetamido and 3363.09 cm –1 of hydroxyl group<br />

indicates the completion of the reaction. Their structure<br />

was further supported by their 1H NMR spectral data<br />

that exhibited three proton of N-methyl signal found<br />

at δ value 2.85, where as aromatic proton signal shows<br />

the δ value 6.45–7.5. Subsequent purifi cation yielded<br />

fi nal compounds in moderate to higher yields. Physical<br />

data of the synthesized compounds are listed in Table<br />

1 and Table 2. Some of these compounds have shown<br />

antibacterial and potent antifungal agents.<br />

64 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


CONCLUSION<br />

Girish Bolakatti et al.: Synthesis and antimicrobial activity of 4-Hydroxy-1-methyl/phenyl-3- (Substituted anilinoacetyl) quinolin-2(1H)-one<br />

4-Hydroxy-1-methyl/phenyl-3-(Substituted<br />

anilinoacetyl)quinolin-2(1H)-one compounds were<br />

synthesized and evaluated for antimicrobial activity<br />

against Staphylococcus aureus, Bacillus substilis Escherichia coli,<br />

Pseudomonas aeruginosa, Candida albicans and Asparagillus<br />

niger. Among synthesized novel compounds 3a, 3b, 3d,<br />

4a, 4c shows promising antibacterial activity as compared<br />

to Ciprofl oxacin (100 μg/ml), however most of them<br />

3a, 3b, 3d, 3e, 4a, 4b, 4d, 4e showed potent antifungal<br />

activity as compared to Fluconazole (100 μg/ml).<br />

Suitable molecular modifi cation of these compounds<br />

may generate potent antimicrobial agents in future.<br />

REFERENCE<br />

1. Alireza F, Saeed E, Masood M, Mohammad HM, Abbas S. Synthesis<br />

and antibacterial activity of N-[2-(5-bromothiophen-2-yl)-2-oxoethyl] and<br />

N-[2-(5-bromothiophen-2-yl)-2-oximinoethyl] derivatives of piperazinyl<br />

quinolones. Bioorg Med Chem Lett 2005; 15: 4536–9.<br />

2. Thomas K. The pyrano route to 4-hydroxy-2-quinolone and 4-hydroxy-<br />

2-pyridones. IL farmaco 1999; 54:309–15.<br />

3. Jayashree BS, Seeja T, Yogendra N. Design and synthesis of<br />

2-quinolones as antioxidants and antimicrobials: a rational approach.<br />

Med Chem Res 2010;19:193–209.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 65


Girish Bolakatti et al.: Synthesis and antimicrobial activity of 4-Hydroxy-1-methyl/phenyl-3- (Substituted anilinoacetyl) quinolin-2(1H)-one<br />

4. El-Dine SA, El-Khawass SM. Synthesis of 4-[(2-alkylaryl or arylalkyl)<br />

amino-1,3,4-thiadiazol-5-yl]-2-synthesized quinolines and compounds<br />

reasonable antifungal and antimicrobial activity. Pharmazie 1979;<br />

34:537–8.<br />

5. Joseph P, Charles B, Gladys D, Trevor V. Refomatsky reactions<br />

with N-arylpyrrolidine-2-thiones: Synthesis of tricyclic analogues of<br />

quinolone antibacterial agents. Tetrahedron 2001;57:9635–48.<br />

6. Anastasia D, Dionysia B, Kyriakos CP, Maria K, Giorgos A, Georgia<br />

M. Design and synthesis of novel quinolinone-3-aminoamides and<br />

their α-lipoic acid adducts as antioxidant and anti-infl ammatory agents.<br />

J Med Chem 2007;50:2450–8.<br />

7. Joseph S, Francis D. Aryl-2-quinolone derivatives having in vivo and<br />

in vitro antitumour activity. J Med Chem 2002;45:2543.<br />

8. Qun Li, Keith WW, Weibo W, et al. Design, synthesis, and activity of<br />

achiral analogs of 2-quinolones and indoles as non-thiol farnesyltransferase<br />

inhibitors. Bioorg Med Chem Lett 2005;15:2033–9.<br />

9. Gisela CM, Mariela B, Ana MB, Silvia EA. Evaluation of antiparasitic,<br />

antituberculosis and antiangiogenic activities of 3-aminoquinolin-2-one<br />

derivatives. J Chil Chem Soc 2006; 51:859–63.<br />

10. Anquetin G, Greiner J, Mahmoudi N, et al. Design, synthesis and activity<br />

against Toxoplasma gondii, Plasmodium spp., and Mycobacterium<br />

tuberculosis of new 6-fl uoroquinolones. Eur J Med Chem 2006;41:<br />

1478–93.<br />

11. Alexandra A, Veziris N, Cambau E, Truffot-Pernot C, Jarlier V, Fisher LM.<br />

Novel Gyrase Mutations in Quinolone-Resistant and Hyper susceptible<br />

Clinical Isolates of Mycobacterium tuberculosis: Functional Analysis of<br />

Mutant Enzymes. Antimicrob Agents Chemother 2006;50:04–112.<br />

12. Cheng AF, Yew WW, Chan EW, Chin ML, Hui MM, Chan RC. Multiplex<br />

PCR amplimer conformation analysis for rapid detection of gyrA<br />

mutations in fl uoroquinolone-resistant Mycobacterium tuberculosis<br />

clinical isolates. Antimicrob Agents Chemother 2004; 48:596.<br />

13. Ginsburg AS, Grosset JH, Bishai WR. Fluoroquinolones, tuberculosis,<br />

and resistance. Lancet Infect Dis 2003;3:432–42.<br />

14. Roschger P, Fiala W, Sradlabauer W. Nucleophilic substitution and<br />

ringclosure reactions of 4-chloro-3-nitro-2-quinolones. J Heterocycl<br />

Chem 1992;29:225–31.<br />

15. Fairbrother RW, Martyn G. The Disc Technique for Determining<br />

Sensitivity to the Antibiotics. J Clin Pathol 1951;4:374–77.<br />

16. Gould JC, Bowie JH. The determination of bacterial sensitivity to<br />

antibiotics. Edinb med J 1952;59:178–99.<br />

17. Handan A, Oznur A, Seher B, Gulten O, Melten U, Dilek S. Synthesis<br />

of mannich bases of some 2,5-disubstituted-4-thiazolidinones and<br />

evaluation of their antimicrobial activities. Turk J Chem 2005;29:425–35.<br />

18. Wolfson J, Hooper DC. Fluoroquinolone Antimicrobial agents. Clini<br />

Microbiol Rev 1989;2(4): 378–24.<br />

66 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Evaluation of gastroprotective ability of<br />

Amorphophallus paeoniifolius corms against<br />

indomethacin induced gastric ulcers<br />

H.N. Nataraj 1 , R.L.N. Murthy 1 , Ramachandra Setty 2<br />

1 Dept. of Pharmacognosy, T.V.M. College of Pharmacy, Bellary<br />

2 Dept. of Pharmacology, Govt. College of Pharmacy, Bangalore<br />

ABSTRACT:<br />

Peptic ulcer is regarded as a multifactorial gastrointestinal disorder in its pathophysiology including free radical<br />

generations. Thereby free radical scavengers can play an important role in such diseases. The methanolic extract<br />

of Corms of Amorphophallus paeoniifolius exhibited remarkable anti-oxidant activity in various In-vitro models.<br />

Further, preliminary phytochemical screening revealed the presence of polyphenolic compounds; fl avonoids and<br />

tannins which are known to possess anti-ulcer activity. In light of these fi nding, it was under taken to investigate the<br />

gastroprotective activity of methanolic extract gainst NSAID–Indomethacin (30 mg/kg p.o.) induced gastotoxicity<br />

in Wistar albino rats wherein the animals were orally administered with two different doses of test extract<br />

(250 and 500 mg/kg b.w.) or with reference drug Lansoprozole (8 mg/kg p.o.). Animals were analyzed for Ulcer<br />

score, and in vitro estimation of GSH and LPO. Extract showed signifi cant (p


H.N Nataraj et al.: Evaluation of gastroprotective ability of Amorphophallus paeoniifolius corms against indomethacin induced gastric ulcers<br />

antisecretory ctivity can prove effectively in PUD<br />

(Peptic Ulcer Disease). Acid neutralization is being recognized<br />

as effective treatment for many centuries ago,<br />

but with the understanding of pathogeneses of PUD<br />

the treatment has become more effective. A number<br />

of antiulcer drugs by various mechanisms like gastric<br />

anti-secretory drugs, H 2 -receptors antagonists, antimuscarine<br />

drugs, proton pomp inhibitors and mucosal protective<br />

agents are in most common usage as a remedy<br />

for peptic ulcer.<br />

In ancient system of medicine, herbal preparations are<br />

being used for treating duodenal ulcers. Presently, a large<br />

section of the world’s population relies on traditional<br />

and medicinal herbs due to their less cost and easy<br />

access. 3 In the Indian Pharmaceutical Industry, antacids<br />

and antiulcer share 6.2 million rupees and occupy 4.3%<br />

of the market share. 4 Some of the phytoconstituents<br />

known to possess antiulcer activity include fl avonoids,<br />

saponins, tannins, gum and mucilages.<br />

Amophophallus paeoniifolius (Dennst.) Nicolson (Family:<br />

Araceae) is a perennial herb with underground tuber<br />

commonly known as ‘Suran’ in Hindi. The plant is widely<br />

distributed in India, Bangladesh, and Africa. 5–6 These<br />

tuberous roots of the plant have been used traditionally<br />

for the treatment of piles, abdominal pain, tumours,<br />

enlargement of spleen, asthma and rheumatism. 5,6 The<br />

tuberous roots have been reported to possess tonic,<br />

stomachic and appetizer properties. 6,7<br />

It was revealed that the corms are reported in the<br />

management of Haemorroids, 8 Obesity; 9 evaluated<br />

for Antiprotease, 10 Analgesic, 11 Immunomodulatory, 12<br />

Anthelmintic, 13 Hepatoprotective 14 and Cytotoxic<br />

activity. 15<br />

Previously we have reported that the methanolic extract<br />

of corms Amophophallus paeoniifolius (MECAP) found to<br />

be containing appreciable quantities of phenolics and<br />

fl avonoids as upon their quantitative estimations, 16 based<br />

on which the possible free radical scavenging potential<br />

by various models of MECAP was anticipated. 17<br />

The present investigation was aimed to evaluate the<br />

gastroprotective potential of Amophophallus paeoniifolius<br />

corms against indomethacin-induced gastric ulcer in<br />

albino rats in pursuit of newer gastro protectant.<br />

MATERIALS AND METHODS<br />

Plant material and preparation of extract<br />

The corms of Amorphophallas paeoniifolius were collected<br />

from cultivated lands from Hassan district of Karnataka<br />

and authenticated by Dr. Kotresh Botany department,<br />

Karnataka University, Dharwad. The voucher specimens<br />

of these plants and tubers were preserved in the<br />

herbarium of the pharmacognosy department of this<br />

institution.<br />

The air dried powdered plant material-corm was<br />

extracted with methanol in soxhelt extraction apparatus<br />

as methanol is one of best solvents for phenolics and<br />

it gave more quantitative colour reaction for phenolics<br />

than hydroalcoholic extract. The methanolic extract was<br />

fi ltered and evaporated to dryness in vacuum (at 35ºC &<br />

0.8Mpa) in a Buchi evaporator, R-114. The dry extract<br />

(MECAP-Methanolic Extract Corms of Amorphophallas<br />

paeoniifolius) was kept in vacuum desiccators until use<br />

and the preliminary phytochemical analysis revealed the<br />

presence of mainly polyphenols, tannins, fl avonoids,<br />

coumarins and triterpenoids.<br />

Animals<br />

Healthy adult Wistar albino rats weighing 200–250 g<br />

were used for the present investigation. They were<br />

housed in clean polyacrylic cages with not more than<br />

four animals per cage and maintained under standard<br />

laboratory conditions (temperature 25 ± 2°C, relative<br />

humidity 55–65%, with dark/light cycle 12/12 h).<br />

They were allowed free access to standard pellet diet<br />

and water ad libitum. The animals were acclimated to<br />

laboratory condition for one week prior to experiment.<br />

All the procedures were performed in accordance with<br />

the institutional Animal Ethics Committee (IAEC)<br />

constituted as per the direction of the Committee<br />

for the Purpose of Control and Supervision of<br />

Experiments of Animals (CPCSEA), under Ministry<br />

of Animal Welfare Division, Govt. of India,<br />

New Delhi. The experiment protocol was approved<br />

by IAEC S.C.S. College of Pharmacy, Harapanahalli,<br />

Karnataka (SCSCP/753/2009-10/Sl No.15).<br />

EXPERIMENTAL<br />

Acute Oral toxicity Studies and selection of dose<br />

Fixed dose method of Organization of Economic<br />

Co-operation and Development (OECD) Guideline<br />

No.423 (Annex 2d) given by CPCSEA18 was adopted for<br />

toxicity studies and it was concluded that the extract was<br />

unclassifi ed w.r.t. Globally Hormonized Classifi cation<br />

system and their LD was infi nite i.e. more than<br />

50<br />

5000 mg/kg. Hence, the doses 500 mg/kg and 250 mg/<br />

kg (1/10th & 1/20th of 5000 mg/kg) were selected for<br />

pharmacological screening of the MECAP.<br />

Antiulcer Activity Studies<br />

Antiulcer activity was evaluated in indomethacin<br />

induced gastric ulcer model according to the method<br />

described by Datta GK et al. 19 In this method, animals<br />

were divided into four groups (each group contains<br />

six rats) and animals were fasted for 36 h prior to the<br />

experiment, but allowed free access to water.<br />

68 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


H.N Nataraj et al.: Evaluation of gastroprotective ability of Amorphophallus paeoniifolius corms against indomethacin induced gastric ulcers<br />

Grouping and Treatment of Animals<br />

The different groups were treated as follows;<br />

Group-I : + ve control water, 10 ml/kg,<br />

P.O. + Indomethacin, 30 mg/kg, P.O.<br />

Group-II : Standard Lansoprazole, 8 mg/kg,<br />

P.O. + Indomethacin, 30 mg/kg, P.O.<br />

Group-III : Methanolic extract 250 mg/kg,<br />

P.O. + Indomethacin, 30 mg/kg, P.O.<br />

Group-IV : Methanolic extract 500 mg/kg<br />

P.O.+ Indomethacin, 30 mg/kg, P.O.<br />

Groups of 6 animals each were pretreated with water<br />

or standard drug or methanolic extract of corm (250,<br />

500 mg/kg P.O.) 30 minutes before the administration<br />

of indomethacin at dose 30 mg/kg orally.<br />

Determination of gastric ulcer index<br />

Four hours later the albino rats were sacrifi ced by an<br />

overdose of anesthetic ether. Subsequently, stomachs<br />

were removed and cut open along the greater curvature<br />

and pinned on a soft board. The number of ulcers in the<br />

glandular portion per stomach were noted and severity<br />

of gastric mucosal lesions scored microscopically with<br />

the help of hand lens (10x) and scoring was done as<br />

fallows. 20<br />

0 = Normal stomach 0.5 = Red coloration<br />

1 = Spot ulcers 1.5 = Haemorrhagic streaks<br />

2 = Ulcer > 3 mm < 5mm 3 = Ulcers > 5mm<br />

Mean ulcer score for each animal was expressed<br />

as ulcer index. The percentage protection was<br />

calculated by using the formula; Percentage protection<br />

= 1–U t /U c × 100 ; Where, U t = Ulcer index of treated<br />

group; U c = Ulcer index of control group (Table 1).<br />

GSH estimation in Indomethacin induced<br />

gastric ulceration<br />

Tissue samples of stomach were homogenized in ice<br />

cold Trichloroacetic acid (1 gm tissue plus 10 ml 10%<br />

TCA) in an ultra turrax tissue homogenizer. Glutathione<br />

measurements were performed using the modifi cation<br />

of the Ellamn procedure. 21 Briefl y, after centrifugation<br />

at 3000 rpm for 10 minutes, 0.5 ml supernatant was<br />

added to 2 ml of 0.3 M disodium hydrogen phosphate<br />

solution. A 0.2 ml solution of dithiobisnitrobenzoate<br />

(0.4 mg/ml in 1% sodium citrate) was added and the<br />

absorbance at 412 nm was measured immediately<br />

after mixing. Percentage increase in OD was directly<br />

proportional to the increase in the levels of Glutathione.<br />

Hence, Percentage increase in OD was calculated by the<br />

formula: Percentage increase = { (Test OD - Control<br />

OD) / Control OD } x 100 (Figure 1).<br />

In vivo lipid peroxidation estimation in gastric<br />

ulceration<br />

The degree of lipid peroxide formation was assayed by<br />

monitoring thiobarbituric reactive substance formation. 22<br />

Combined 1.0 ml of tissue samples of stomach<br />

biological sample (0.1–2.0 mg of membrane protein or<br />

0.10.2μ mol of lipid phosphate) with 2.0 ml of TCA-<br />

TBA-HCl (15% w/v trichloroacetic acid; 0.375% w/v<br />

thiobarbituric acid; 0.25N hydrochloric acid) and mixed<br />

thoroughly. The solution was heated for 1 hr in a boiling<br />

water bath. After cooling, the fl occulent precipitate was<br />

removed by centrifugation at 1000 rpm for 10 mins. The<br />

absorbance of the sample was determined at 535 nm<br />

against a blank that contains all the reagents minus the<br />

lipid. The malondialdehyde concentration of the sample<br />

was calculated by using an extinction coeffi cient of<br />

1.56 × 105 M –1 cm –1 [Percentage inhibition = { (Control<br />

– Test) / Control OD } × 100] (Figure 2).<br />

Statistical analysis<br />

All the results were expressed as mean ± SEM, n = 6.<br />

Statistical analysis was performed with one way analysis<br />

of variance (ANOVA) followed by Tukey-Kramer<br />

Multiple Comparisons Test by using Graph Pad Instat<br />

3.06 Software P value less than < 0.05 was considered<br />

to be statistically signifi cant. * P < 0.05, **


H.N Nataraj et al.: Evaluation of gastroprotective ability of Amorphophallus paeoniifolius corms against indomethacin induced gastric ulcers<br />

Figure 1: Effect of MECAP on tissue GSH levels.<br />

Figure 2: Effect of MECAP on tissue GSH levels.<br />

of ethyl acetate were spotted in the form of bands on<br />

precoated and preactivated aluminium silica gel 60GF 254<br />

HPTLC plates (10 cm × 10 cm width, 0.2 mm thickness-<br />

E-Merk) by means of Linomat IV automatized spray<br />

on band applicator equipped with a 100 μL syringe. The<br />

linear ascending development was carried out in Camag<br />

HPTLC twin trough chamber saturated with mobile<br />

phase consisting of chloroform : ethyl acetate : formic<br />

acid (5:4:1). Subsequent to the development the plates<br />

were dried in a current of air with the help of an air dryer.<br />

Densitometric scanning was performed on Camag TLC<br />

scanner III in absorbance mode at 254 nm (Figure 3).<br />

For references, HPTLC fi nger print analysis of standards<br />

markers viz., Gallic acid and Quercetin were developed<br />

70 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


H.N Nataraj et al.: Evaluation of gastroprotective ability of Amorphophallus paeoniifolius corms against indomethacin induced gastric ulcers<br />

Figure 3: HPTLC Finger print profile after densitometric scan of Sample (EtOAc fraction of MeOH extract) under uv 254 nm (Chloroform:<br />

Ethyl acetate: Formic acid - 5:4:1).<br />

with same solvent system. Densitometric scanning was<br />

performed and R f values were recorded (Table 2).<br />

RESULTS<br />

Effect of MECAP on tissue GSH & LPO levels,<br />

Gastric ulceration<br />

In pylorus ligation induced gastric ulcer there was<br />

marked reduction in the tissue GSH levels in control<br />

group. Treatment with MECAP increased GSH levels<br />

by both the doses 250 and 500 mg/kg to 47.01% &<br />

73.09% respectively and inhibition of lipid peroxidation<br />

generation up to 29.27% and 36.81% respectively in a<br />

dose dependant manner. Besides, MECAP at both<br />

concentrations showed signifi cant (p


H.N Nataraj et al.: Evaluation of gastroprotective ability of Amorphophallus paeoniifolius corms against indomethacin induced gastric ulcers<br />

role in gastric ulceration induced by several types of<br />

stress. ROS also decrease the levels of endogenous<br />

antioxidants, such as GSH, α-tocophenol and ascorbate<br />

and make more prone to oxidative damage. 26 Most of<br />

the world’s population relies on traditional remedies<br />

for its treatment. It is recommended that natural drugs<br />

containing antiulcer, antioxidant and antisecretory<br />

activity can exhibit effectiveness in peptic ulceration.<br />

The well known Phytoconstituents possessing antiulcer<br />

activity includes polyphenols, tannins, fl avonoids, gums<br />

and mucilages, saponins etc.<br />

In the present study indomethacin induced gastric ulcer<br />

model experiment, ulcer index parameter was used<br />

for the evaluation of gastroprotective activity. Oral<br />

administration of indomethacin (30 mg/kg) has induced<br />

ulcers as indicated by higher ulcer index values. Further<br />

it also reduced the gastric tissue glutathione levels<br />

and enhanced the lipid peroxidation. Indomethacin<br />

induced gastric damage particularly due to inhibition of<br />

cycloxygenase pathway of arachidonic acid metabolism<br />

which has a consequence diversion of pathway resulting<br />

in over production of leukotrienes 27,28 and these<br />

leukotrienes have been attributed an important role<br />

in gastrointestinal ulceration. 29 Apart from this, these<br />

agents break the mucosal barrier, provoke an increase in<br />

the gastric mucosal permeability to H + and Na + ions and<br />

drop in the transmucosal potential differences inducing<br />

the formation of erosions and ulcer and in addition<br />

indomethacin markedly decrease gastric mucosal PGE 2<br />

levels causing ulceration. 30<br />

Whereas, the Methanolic extract of corms of<br />

Amorhophallus paeoniifolius (MECAP) signifi cantly reduced<br />

the ulcer score in dose dependent manner compared to<br />

control group. Along with this, it also lowered destructive<br />

lipid peroxidation (LPO) levels and restored back the<br />

protective GSH levels.<br />

Preliminary phytochemical screening of MECAP indicated<br />

the presence of the phytoantioxidants viz., Tannins,<br />

Flavonoids, Sterols, Polyphenolics and Coumarins<br />

which are well known powerful natural antioxidants due<br />

to their electron donating property which either scavenge<br />

the principal propagation free radicals or halt the<br />

radical chain. The HPTLC analysis revealed the presence<br />

of nine phenolics including Gallic acid and Quercetin<br />

in the tested extract.<br />

Besides, various mechanisms have been thought to be<br />

involved in the ulcer production; hence it is not possible<br />

to propose a single mechanism for anti-ulcer effect to a<br />

particular drug or especially for an extract. It has been<br />

demonstrated that the antioxidant compounds could<br />

be active in producing antiulcerogenic effect as these<br />

substances are capable of settling on the membrane<br />

and counteracting lipid peroxidation. 31 Along with<br />

this the fl avonoids present in the test extract not only<br />

exhibit antioxidant activity 32 but also have effects in<br />

gastrointestinal tract, including mucosal protection in<br />

rat colitis. 33 In addition to this, it is being reported that<br />

fl avonoids are able to protect the gastric mucosa against<br />

a variety of ulcerogenic agents via several mechanisms<br />

of action, mainly free radical scavenging and antioxidant<br />

properties, increased mucus production. 34 Thus the<br />

antiulcerogenic effect observed with oral administration<br />

of MECAP could be related to the presence of fl avonoids<br />

and other phytoantioxidants detected in test extract.<br />

However, further studies are required to establish the<br />

exact mode of action and the active principles involved<br />

in its gastroprotection.<br />

CONCLUSION<br />

In conclusion, from the results of the present study, it is<br />

clear that Amorphophallus paeoniifolius corms methanolic<br />

extract has shown signifi cant gastroprotective<br />

activity in indomethacin induced gastric ulceration<br />

animal model. The MECAP at both doses exhibited<br />

signifi cant gastoprotection in dose dependent manner<br />

in comparison to standard Lansoprazole. In addition to<br />

tannins, sterols, fl avonoids, coumarins and nine phenolic<br />

compounds including quercetin and gallic acid (resolved<br />

in HPTLC analysis) present in extract and some other<br />

compounds might have contributed to the protection<br />

offered. Further, evaluation of the said compounds for<br />

gastroprotection is under study to ascertain the claim.<br />

ACKNOWLWDGMENTS<br />

The authors are grateful to principal and management,<br />

T.V.M. College of Pharmacy, Bellary and authorities<br />

of SCS College Pharmacy, Harapanahalli for providing<br />

necessary facilities. We wish to extend our thanks to<br />

Dr. Kotresh also for authentication of plant.<br />

REFERENCES<br />

1. Shenoy AM, Shastry CS, Sridevi B, Gopkumar P. Antiulcer activity of<br />

Naravelia zeylanica leaves extract. J. Pharm Res. 2009;2:1218–20.<br />

2. Kassel N and Munro, Mind And Ulcer, Brit Med J. 1996;3:374–76.<br />

3. Chetterjee TK, Herbal options Calcutta, <strong>Book</strong>s and Allied Pvt. Ltd. 2001,<br />

203–50.<br />

4. Dharmani P and PAtil G, Exploring Indian medicinal plants for antiulcer<br />

ativity. Indian J Pharmacol, 2006;38:95–6.<br />

5. Bhattacharya S, Chironjib banoushadi, Calcutta : Anand Publishing Ltd.<br />

India; 1990;(2), I. 63–9.<br />

6. Ghani A, Medicinal plants of Bangladesh. Dhaka : Asiatic society of<br />

Bangladesh publisher Ltd ; 1998, 77–8.<br />

7. Kirtikar KR, Bsau BD, Indian Medicinal Plants. India : Dehra Dun<br />

publisher Ltd. 1994, (4), II, 2609–10.<br />

8. Vastrad CS, Pakkanavar RV, ‘A herbal formulation in the management,<br />

Haemorroids’, Antiseptic, 99;9:343–4.<br />

9. Li B, Xia J, Wang Y, Xie B, ‘Antiobesity activity of corm of Amophophallus<br />

paeoniifolius. J. Agri Food. Chem., 2002;53:740–5.<br />

10. Pratibha, S, Nambison B and Leelamma, ‘Enzyme inhibiters in tubers<br />

crops’, Plant Food Hum Nutr., 1995;48:247.<br />

11. Shilpi JA, Ray PK, Sarder MM, and Uddin SJ ‘Analgesic activity of<br />

Amorphophallus companulatus tuber’, Fitoteropia, 2005, 367–9.<br />

72 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


H.N Nataraj et al.: Evaluation of gastroprotective ability of Amorphophallus paeoniifolius corms against indomethacin induced gastric ulcers<br />

12. Shastry RA, Mahaden KM, Habbu PV, Patil JP. Immunomodulatory<br />

activity of corm extracts of Amophophallus paeoniifolius (Dennst),<br />

Pharmakine, 2009;1(4):26–33.<br />

13. Ramalingam J, Hima Bindu K, Madhavi B, Ravinder Nath A, David Banji,<br />

‘Phytochemical and Anthelmintic eavaluation of corm Amorphophallus<br />

companulatus’, International <strong>Journal</strong> of Pharma and Bioscinces, 2010,<br />

VI(2), 1–9.<br />

14. Jain S, Dixit VK and Malviya N, ‘Antioxidant and hepatoprotective activity<br />

of Ethanolic and Aqueous extracts of Amorphophallus campanulatus’ ,<br />

Acta poloniac Pharmaceutica, 2009;66(4):423–428.<br />

15. Angayarkanni J, Ramkumar KM, Poornima T, Priyadarshini U, Cytotoxic<br />

activity of Amorphophallus paeoniifolius tuber extract, American-<br />

Euresian J Agri Environ Sci, 2007;2:395–8.<br />

16. Nataraj HN, Murthy RLN, Ramachandra setty S, In vitro quantifi cation<br />

of Flavonoids And Phenolic content of–Suran, Int. J. ChemTech Res,<br />

2009;1(4):1063–1067.<br />

17. Nataraj HN, Murthy RLN, Ramachandra setty S, ‘In vitro antioxidant<br />

and free radical scavenging potential of Amorphophallus paeoniifolius’,<br />

Oriental journal of chemistry, 2008;24(3):895–902.<br />

18. Mrs. Prema Veeraraghavan. Expert consultant, CPCSEA, OECD<br />

guideline No. 423, Oct. 2000.<br />

19. Datta GK, Sairam K, Priyambada S, Debnath PK, Goel RK.<br />

Antiulerogenic activity of Satavaru mandur- An ayurvedic herbomineral<br />

Preparation. Indian J Expl Biol 2002;40:1173–7.<br />

20. Kulkarni SK. Hand book of experimental pharmacology, New Delhi,<br />

Vallabhah prakashan ; 1999, 128–131.<br />

21. Aykae G, Vysal M, Yalein AS, Kocak-Toker N, Sivas A, Oz H. The effect<br />

of chronic ethanol ingestion on hepatic lipid peroxide, Glutathione,<br />

glutathione peroxidase and glutathione transferase in rats. Toxicology<br />

1985;36:71–6.<br />

22. John Buege A, Steven Aust D, Microsomal lipid peroxidation. London:<br />

Moury Kleiman Co., 1978, 302.<br />

23. Repetto MG and Liesuy SF, Antioxidant properties of natural compounds<br />

used in popular medicine for gastric ulcers, Brazil J Med Biol Res, 2002;<br />

35(5):523–524.<br />

24. Pihan G, Rogers C, Szabo S, Free radicals and lipid peroxidation in<br />

ethanol and aspirin induced gastric mucosal injury, Digestive Diseases<br />

Sci, 1987;32(12):1395–1399.<br />

25. Vaananann PM, Medding JB and Wallace JL, Role of oxygen derived<br />

free radicals in indomethacin-induced gastric injury, Amer J Physiol,<br />

1991;261:470–475.<br />

26. Phull PS, Green CJ, Jaycna MRA, A radical review of the stomach; the<br />

role of oxygen derived free radicals and antioxidants in gastroduodenal<br />

diseases, Eur J Gastroenterol Hepatol, 1995;7:265–271.<br />

27. Pihan G, Rogers C, Szabo S., Vascular injury in acute gastric mucosal<br />

damage, mediatory role of leukotrienes. Dig. Dis. Sci 1988;33:<br />

625–632.<br />

28. Rainsford KD. Gastric ulcerogenicity of non-steroidal anti-infl ammatory<br />

drugs in mice with mucosa sensitized by anolinmimetic treatment.<br />

J. Pharmacol 1978;39:669–672.<br />

29. Wallace JL, McKnight, G.W, Keenan CM., Byles, NI, MacNaughton WK.<br />

Effects of leukotrienes on susceptibility of the rat stomach to damage<br />

and investigation of the mechanism of action. Gastroenterology<br />

1990;98:1178–1186.<br />

30. Whittle BJ. Temporal relationship between cyclooxygenase inhibition,<br />

as measured by prostacyclin biosynthesis and the gastrointestinal<br />

damage induced by indomethacin in rat. Gastroenterology 1981;80(1):<br />

94–98.<br />

31. Halliwell B. Antioxidants in human health and disease. Annu. Rev. Nutr<br />

1996;16:33–50.<br />

32. Magnani L, Gaydou EM, Hubaud JC. Spectrophotometric, measurement<br />

of antioxidant properties of fl avones and fl avonols agaisnt superoxide<br />

anion. Analytica Chim. Acta. 2000;411:209–216.<br />

33. Gálvez J, Cruz T, Crespo E, Ocete MA, Iorente MD, Sánchez de Medina<br />

F, Zarzuelo, A. Rutoside as mucosal protective in acetic acid-induced<br />

rat colitis. Planta Med 1997;63:409–414.<br />

34. Shay M, Kamarov SA, Fels D, Meraaze D, Grueinstein H, Siplet H. A<br />

simple method for the uniform production of gastric ulceration in the rat.<br />

Gastroenterology 1945;5:43–61.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 73


Immunomodulatory activity of methanolic<br />

extracts of Pongamia glabra Vent. seeds and<br />

bark in cyclophosphamide induced mice<br />

Sanjeev Heroor 1 , Arunkumar Beknal 1 , Nitin Mahurkar 2<br />

1 Dept. of Pharmacognosy and Phytochemistry, HKES’s MTR institute of Pharmaceutical Sciences, Gulbarga, Karnataka, India<br />

2 Dept. of Pharmacology, HKES’s MTR institute of Pharmaceutical Sciences, Gulbarga, Karnataka, India<br />

ABSTRACT<br />

Immune activation is an effective as well as a protective and novel approach against emerging infectious<br />

diseases. Traditionally Pongamia glabra Vent claimed to cure infectious diseases needs scientifi c validation as<br />

immunomodulatory agent. Methanolic extracts of seeds and barks at the doses of 250 mg/kg and 500 mg/kg<br />

(per oral) of Pongamia glabra Vent. were studied for the assessment of immunomodulatory activity on<br />

cyclophosphamide induced immunosuppression in mice. The activity was assessed by determining the RBC,<br />

Hb%, platelet, total WBC and differential counts. Methanolic extracts of seeds and barks of Pongamia glabra<br />

Vent. showed dose dependent highly signifi cant counteracting effect (p


Sanjeev Heroor et al.: Immunomodulatory activity of methanolic extracts of Pongamia glabra Vent. seeds and bark in cyclophosphamide induced mice<br />

Karanja, a tree found all over India bearing imparipinnate<br />

leaves and pinkish white colored fl owers. 8 The seeds of<br />

the plant reported to contain fi xed oil and traces of<br />

essential oil. Bark contains a bitter alkaloid, resin and<br />

mucilage. 9 The ancient folklore claims the uses of the<br />

seeds to treat rheumatic joints and anti-paracytic. Bark is<br />

astringent and powdered seeds are used as febrifuge and<br />

tonic. 10 The present study was aimed at screening of seeds<br />

and bark methanolic extracts of Pongamia glabra Vent.<br />

for immunomodulatory activity in cyclophosphamide<br />

induced immunosuppressed albino mice.<br />

MATERIALS AND METHODS<br />

Plant material<br />

Pongamia glabra Vent. seeds and bark were collected from<br />

local areas of North Karnataka and a voucher specimen<br />

has been deposited at the departmental herbarium. The<br />

plant was authenticated by Dr. Srinathrao, Prof. and<br />

HOD, Dept. of Botany, Gulbarga University, Gulbarga<br />

and Ref. No. GUG/BOT/Herbarium/2008–09/09. The<br />

mentioned parts of the plant were dried and pulverized<br />

to particle size (#) 40 and then were fi rst defatted with<br />

petroleum ether (40–60°C) and extracted with methanol<br />

by continuous hot percolation method using Soxhlet<br />

apparatus at 40°C for 48 h to obtain methanolic extracts<br />

of seeds and bark of the plant respectively. The fi ltrates<br />

of the extracts were concentrated to dryness at 40°C<br />

under reduced pressure in a rota fl ash evaporator. The<br />

yields of the methanolic extracts of seeds and barks were<br />

43.16 gm (30.59%w/w) and 37.76 gm (22.19%w/w)<br />

respectively.<br />

Preliminary Phytochemical Studies<br />

The methanolic extracts of seeds and bark were<br />

subjected for preliminary qualitative chemical tests<br />

and the presence of major phytoconstituents were<br />

confi rmed by Thin Layer Chromatography (TLC)<br />

studies11,12 (Table 1).<br />

Thin Layer Chromatography studies<br />

• TLC profi le for fl avonoids<br />

Solvent system: ethyl acetate: formic acid: glacial<br />

acetic acid: water<br />

(10:1.1:1.1:2.6).<br />

Detection: UV 365 (blue fl uorescent spots).<br />

• TLC profi le for alkaloids<br />

Solvent system: toluene: ethyl acetate: diethylamine<br />

(7:2:1)<br />

Detection: wagner’s reagent (brown colored spots).<br />

• TLC profi le for steroids<br />

Solvent system: petroleum ether: acetone (7:3).<br />

Detection: anisaldehyde: sulphuric acid reagent (pink<br />

to red colored spots)<br />

• TLC profi le for saponins<br />

Solvent system: chloroform: glacial acetic acid:<br />

methanol: water (6.4:3.2:1.2:0.8).<br />

Detection: anisaldehyde: sulphuric acid reagent (pink<br />

colored spots).<br />

Animals<br />

Swiss albino mice of either sex, weighing 25–30 g<br />

housed in standard conditions of temperature, humidity<br />

and light were used. They were fed with standard rodent<br />

diet and water ad libitum. The study was approved by<br />

Institutional Animal Ethical Committee, Ref. No.<br />

HKECOP/IAEC/45/2011–12.<br />

Acute Toxicity Studies<br />

Acute toxicity studies were conducted as per OECD<br />

guideline by 425 method. 13 The animals did not show<br />

any mortality at the dose of 5000 mg/kg and hence its<br />

1/10th dose i.e. 500 mg/kg and 1/20th dose i.e. 250 mg/<br />

kg were used as the therapeutic doses for the methanolic<br />

extracts of the study.<br />

Test Samples<br />

Weighed quantities of test extracts were suspended<br />

in 1% sodium carboxy methyl cellulose to prepare a<br />

suitable dosage form. 14 The control animals were given<br />

an equivalent volume of sodium CMC vehicle.<br />

Drugs<br />

Cyclophosphamide was used as a standard immunosuppressant,<br />

Cycloxan ® (Biochem–pharmaceutical<br />

indus tries Ltd., Mumbai) containing 200mg – cyclosphosphamide<br />

, was procured from the market and dilutions<br />

were made using sterile water for injection as mentioned<br />

on the label of the marketed product.<br />

Cyclophosphamide Induced Myelosuppression15 Animals were divided into six groups of six animals<br />

each. Group I served as control group and received<br />

Table 1: Thin Layer Chromatography (TLC) results of Pongamia glabra Vent. seeds and bark methanolic extracts<br />

Flavonoids Alkaloids Steroids Saponins<br />

S.No. Extracts No. of spots Rf value No. of spots Rf value No. of Spots Rf Value No.of spots Rf value<br />

1 Seed Ext. 2 0.59, 0.63 2 0.89, 0.80 2 0.61, 0.76 2 0.58, 0.65<br />

2 Bark Ext. 2 0.62, 0.71 2 0.82, 0.53 4 0.64,0.72, 0.78, 0.88 2 0.72, 0.87<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 75


Sanjeev Heroor et al.: Immunomodulatory activity of methanolic extracts of Pongamia glabra Vent. seeds and bark in cyclophosphamide induced mice<br />

the vehicle (1% sodium CMC) for 13 days. Group<br />

II (Cyclophosphamide group) received the vehicle<br />

(1% sodium CMC) for a period of 13 days and on 11th,<br />

12th and 13th days was injected with cyclophosphamide<br />

(30 mg/kg intraperitonial). Groups III and IV were<br />

administered methanolic extracts of seeds of the plant<br />

at the doses of 250 mg/kg and 500 mg/kg (per oral)<br />

daily for 13 days respectively. Similarly Groups V and<br />

VI were administered methanolic extracts of bark of<br />

the plant at the doses of 250mg/kg and 500mg/kg (per<br />

oral) daily for 13 days respectively. The groups III, IV, V<br />

and VI were injected with cyclophosphamide (30 mg/kg<br />

intraperitonial) on the 11th, 12th and 13th days, 1 hour<br />

after the administration of the Respective oral treatment.<br />

Blood samples were collected on 14th day of the<br />

experiment by retro orbital puncture and hematological<br />

parameters were studied for RBC, Hb%, platelets, total<br />

WBC counts and differential leucocytes counts (DLC).<br />

Statistical Analysis<br />

Data were expressed as mean ± SEM and differences<br />

between the groups were statistically determined by<br />

analysis of variance (ANOVA) followed by Dunnet’s<br />

test. p-values


Sanjeev Heroor et al.: Immunomodulatory activity of methanolic extracts of Pongamia glabra Vent. seeds and bark in cyclophosphamide induced mice<br />

Immune activation is an effective as well as protective<br />

approach against emerging infectious diseases. 17<br />

Immunomodulatory activity of methanolic extract of<br />

seeds and bark of Pongamia glabra Vent. was explored<br />

by evaluating their effects on cyclophosphamide<br />

induced myelosuppression in mice at 2 dose levels of<br />

250 mg/kg and 500 mg/kg (per oral) Results of the<br />

study revealed the dose dependent counteracting effect<br />

of the extracts to the cyclophosphamide induced bone<br />

marrow activity suppression i.e. myelosuppression, as<br />

indicated by increase in RBC, total WBC platelet counts,<br />

Hb% and DLC in the extract treated groups (Group III,<br />

IV, V and VI), when compared to cyclophosphamide<br />

treated group (Group II). However the counteracting<br />

activity to reduction in blood cell counts of the seeds<br />

extract was more than that of the bark extract at the<br />

mentioned doses, but both extracts showed similar level<br />

of signifi cance of activity. The potentiated activity of<br />

seed extract may be due to presence of excess number<br />

of fl avonoids and steroids in the seed extract. The<br />

results indicate modulation of bone marrow activity,<br />

viz., – suppression when used cyclophosphamide alone<br />

and stimulation to counteract the cyclophosphamide<br />

induced myelosuppression in pretreated methanolic<br />

extract groups of seeds and barks of Pongamia<br />

glabra Vent.<br />

CONCLUSION<br />

From the phytochemical investigation, it was found<br />

that the major chemical constituents of the methanolic<br />

extracts of seeds and barks were steroids, saponins,<br />

tannins, fl avonoids, alkaloids, proteins and carbohydrates.<br />

Saponins are either triterpenoid or steroidal glycosides<br />

proven as important phytoconstituent with different<br />

pharmacological activities such as antiallergic,<br />

cytotoxic, antitumour, antiviral, immunomodulating,<br />

antihepatotoxic, and antifungal activities. Recently three<br />

diosgenyl saponins isolated from Paris polyphylla were<br />

reported for immunostimulating activity. 18 Tannins are<br />

also known to possess immunostimulating activites. The<br />

well known ayurvedic formulation, Triphala contains<br />

Terminalia chebula, Terminalia belerica and Emblica offi cinalis,<br />

which are rich in tannins reported for immunostimulating<br />

activity. 19 Hence the collective presence of steroids,<br />

saponins, tannins and fl avonoids in the methanolic<br />

extracts would be attributed for immunostimulating<br />

activity. However this is a preliminary research work<br />

and the precise mechanism(s) of immunomodulatory<br />

action infl uenced by potent bio-active constituents of<br />

methanolic extracts of seeds and bark of Pongamia glabra<br />

Vent. against cyclophosphamide induced immunesuppression<br />

needs to be investigated.<br />

ACKNOWLEDGMENT<br />

Authors are thankful to authorities of HKE Society and<br />

MTR Institute of Pharmaceutical sciences, Gulbarga,<br />

Karnataka India, for providing necessary facilities to<br />

carry out the study.<br />

REFERENCES<br />

1. Charak Samhita (Trans.). Shree Gulab Kunvera Ayurvedic Society,<br />

Jamnagar, India. 1949; 249–50.<br />

2. Patwardhan B, Kalbag D, Patki P. S, Nagasampagi B.A. Search of<br />

Immunomodulatory agents: a review. Indian Drugs 1990;28(2):56–63.<br />

3. Wagner H. Immunomodulatory agents. Proceedings of the Alfred<br />

Benzon Symposium. 1983:20;559.<br />

4. Makare N, Bodhankar S, Rangari V. Immunomodulatory activity of<br />

alcoholic extracts of Mangifera indica Linn in mice. J. Ethnopharmacol.<br />

2001;78:133–7.<br />

5. Baumann F, Preiss R. Cyclophosphamide and related anticancer drugs,<br />

B. chrmoatogr. B. Biomed Sci. Appl.2001;764:173–92.<br />

6. Goodman J. W. In : Stites D. P., Terr. A.I., Parslow T. G. (Eds.), The<br />

Immune response in Basic and Clinical Immunology. 8th Edn. NJ:<br />

Prentice-Hall, Engle wood Cliffs; 1994; 40–7.<br />

7. Angulo I., Jimenez M.B., Garcia Bustos J.F, Gargallo D. Candida albicans<br />

infection enhances immunosuppression induced by cyclophosphamide<br />

by selective priming of suppressive myeloid progenitors. cell immunol<br />

2002:218;46–58.<br />

8. Kirtikar K. R, Basu B.D. Indian Medicinal Plants, Oriental Enterprises.<br />

Dehradun, 2nd Ed. 1984;I:830–32.<br />

9. Nadkarni K. M. Indian Materia Medica, Popular Prakashan.1996;<br />

I: 1001–04.<br />

10. Hartwell J. L. Plants used against cancer. A Survey, Lloydia.1967–1971;<br />

30–34.<br />

11. Khandelwal K.R. Practical Pharmocognosy Techniques and<br />

Experiments. 10th Ed. Pune: Nirali Prakashan; 2003; 149–58.<br />

12. Hildebert Wagner, Sabine Bladt. Plant Drug Analysis-A Thin layer<br />

Chomoatography Atlas, Springer-verlag. 2nd Ed. New York: Berlin<br />

Heidelberg; 2001; 1–3, 195–197, 305–206.<br />

13. Committee for the purpose of control and supervision of Experimental<br />

Animals (CPCSEA), OECD Guidelines for the testing of Chemicals,<br />

revised draft guidelines 425: Acute oral toxicity-Acute toxic class method,<br />

revised document. India: Ministry of Social Justice and Empowerment;<br />

2008, No.26.<br />

14. Satpute K. L, Jadhav M. M, Karodi R. S, Patil M.J. Immunomodulatory<br />

activity of fruits of Randia dumetorum Lamk. J. Pharmocog. and<br />

Phytother. 2009;1:1–5.<br />

15. Manjarekar P. N, Jolly C.L, Narayan S. Comparative studies of<br />

immunomodulatory activity of Tinospora cordifolia and Tinopora<br />

sinensis. Fitoterpia 2001;71:254–7.<br />

16. Pelczar M.J, Chan E.C.S, Krieg N. R. Microbiology, 5th Ed. New Delhi:<br />

Tata Mcgraw Hill; 1990; 703–15.<br />

17. Hackett C J. Allergy. Clin. Immunol. 2003;112:686–94.<br />

18. Xiu-feng Z, Yan C, Jiajun H. Ya-Zhou Z, Zhou N. Ya-Lin T, Yang L.<br />

Immunostimulating properties of diosgenyl saponins isolated from Paris<br />

polyphylla. Bioorganic and Med. Chem. Letters 2007;7:255–9.<br />

19. R Srikumar, Parthasarathy N.J, Sheeladevi R. Immunomodulatory<br />

activity of Triphala on Neutrophil Functions. Biol. Pharm. Bull. 2005;<br />

28(8):1398–1403.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 77


Simultaneous fi rst derivative UV<br />

spectrophotometric estimation of<br />

ramipril and olmesartan<br />

Santosh R. Karajgi, Simpi C.C., Kalyane N.V.<br />

Department of Pharmaceutical Chemistry, BLDEA’s College of Pharmacy, BLDEA University Campus, Solapur Road,<br />

Bijapur, Karnataka, India<br />

ABSTRACT<br />

A simple, precise and economical procedure for the simultaneous estimation of Olmesartan Medoxomil and<br />

Ramipril in tablet formulation has been developed. Olmesartan and Ramipril are antihypertensive agents belonging<br />

to category of angiotensin-converting enzyme inhibitor. The present study involves the simultaneous estimation<br />

done by fi rst derivative UV Spectrophotometric method using Shimadzu 1700 spectrophotometer. Olmesartan<br />

has zero crossing point at 240 nm in methanol and Ramipril has zero crossing point at 246 nm in methanol.<br />

Both these drugs obey Beer’s law in the concentration range employed for the present method. The result<br />

of analysis has been validated statistically by recovery studies. The slope and intercept for Olmesartan were<br />

0.0364 and 0.0078 and for Ramipril were 0.0010 and –0.0001 respectively as determined by the method of<br />

least squares.<br />

INTRODUCTION<br />

Ramipril is a prodrug belonging to the<br />

class of ACE inhibitors used to treat<br />

hypertension and congestive heart failure<br />

where as Olmesartan is an ARB used<br />

to treat high blood pressure. Both ACE<br />

inhibitors and ARBs are widely used in<br />

renal failure patients in the treatment of<br />

hypertension, left ventricular dysfunction,<br />

and diabetic nephropathy. Their effi cacy<br />

in these conditions is well established, and<br />

generally both classes of drugs are well<br />

tolerated, with a low incidence of side<br />

effects. 1 Combination of ACE inhibitors and<br />

ARBs is proved to be a useful combination<br />

therapy for the treatment of ischemic heart<br />

diseases. 2<br />

Review of literature revealed that there<br />

are very few methods reported for the<br />

estimation of Ramipril and Olmesartan<br />

individually and in combined dosage<br />

forms, 3–26 no derivative spectrophotometric<br />

method has been so far reported for<br />

simultaneous estimation of these drugs in<br />

combined dosage forms. Therefore, the<br />

aim and objective of present study were<br />

to develop a fi rst derivative UV-Visible<br />

spectrophotometric analytical method for<br />

the estimation of Ramipril and Olmesartan<br />

in bulk and formulated dosage form in<br />

combination without prior separation and<br />

to establish a simple, sensitive, standard,<br />

reproducible method for the quality control<br />

of Ramipril and Olmesartan.<br />

MATERIALS AND METHODS<br />

Materials<br />

Ramipril and Olmesartan were gift samples<br />

from Ajantha Pharmaceuticals, Aurangabad.<br />

The commercial formulations were<br />

purchased from local market. Methanol<br />

A.R. Grade (Qualigens, Fine chemicals)<br />

was used as the solvent.<br />

Research Ar cle<br />

Received Date : 27-08-2011<br />

Revised Date : 06-02-2012<br />

Accepted Date : 08-02-2012<br />

DOI: 10.5530/rjps.2012.1.11<br />

Address for<br />

correspondence<br />

Santosh R. Karajgi<br />

Department of Pharmaceutical<br />

Chemistry, BLDEA’s College of<br />

Pharmacy, BLDEA<br />

University Campus,<br />

Solapur Road, Bijapur,<br />

Karnataka, India<br />

Email: santosh.karajgi@gmail.<br />

com<br />

Mobile: +919739619395<br />

www.rjps.in<br />

78 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Equipments<br />

Santosh R. Karajgi et al.: Simultaneous First Derivative UV Spectrophotometric Estimation of Ramipril and Olmesartan<br />

Shimadzu spectronic 1700 double beam UV-visible<br />

spectrophotometer with 1 cm matched quartz cells was<br />

used for all the absorbance measurements.<br />

Method<br />

The solutions of Ramipril and Olmesatan were<br />

prepared separately in methanol at a concentration<br />

range of 10 μg/ml. Both these solutions were scanned<br />

in the wavelength range of 200 nm to 300 nm. Data<br />

were recorded at the wavelength interval of 1 nm in the<br />

derivative mode to obtain fi rst order derivative spectra<br />

at N=2. After examining the overlain fi rst derivative<br />

spectra, wavelengths for analysis were selected where<br />

one drug showed zero crossing and the other drug<br />

showed substantial absorbance. The wavelength selected<br />

for Ramipril analysis was 240 nm where Olmesartan<br />

has zero absorbance and the wavelength selected for<br />

Olmesartan analysis was 246 nm where the absorbance<br />

of Ramipril is zero. The overlain derivative spectra of<br />

the drugs are given in the Figure 1.<br />

Linearity<br />

Standard stock solutions of Ramipril and Olmesartan<br />

were prepared by dissolving 100 mg each of standard<br />

drug samples in 100 ml volumetric fl asks separately<br />

and the volume was made up with methanol to<br />

get a concentration of 5 – 50 μg/ml for each drug.<br />

Figure 1: Zero crossing points of Ramipril and Olmesartan.<br />

The absorbances of derivative spectra were measured<br />

at 240 nm and 246 nm for Ramipril and Olmesartan<br />

respectively. Five replicate analyses were carried out.<br />

Absorbance against concentrations were plotted to<br />

obtain the calibration graph. Both the drugs obeyed<br />

Beer’s law with the above concentration range with<br />

the R 2 value of 0.9994 and 0.9996 for Ramipril and<br />

Olmesartan respectively (Figure 2 and 3).<br />

Analysis of marketed formulation<br />

Twenty tablets were weighed accurately and powdered.<br />

An accurately weighed quantity of powder equivalent<br />

to 20 mg of Ramipril and 20 mg of Olmesartan was<br />

taken in a 50 ml volumetric fl ask and dissolved in<br />

methanol. The volume was made up with methanol so<br />

that the theoretical concentrations of both drugs were<br />

10μg/ml and the concentrations were determined at<br />

240 nm for Ramipril and 246 nm for Olmesartan by<br />

using calibration graph. (Table 1). The overlain spectra<br />

of synthetic mixture, Ramipril bulk drug, Olmesartan<br />

bulk drug and combined tablet formulation of Ramipril<br />

and Olmesartan is given in the Figure 4.<br />

Recovery Studies<br />

To determine the accuracy of the method, recovery<br />

study was performed using the method of standard<br />

addition. To the pre- analyzed marketed formulation<br />

powder of combined Ramipril and Olmesartan, diluted<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 79


Santosh R. Karajgi et al.: Simultaneous First Derivative UV Spectrophotometric Estimation of Ramipril and Olmesartan<br />

Table 1: Analysis of marketed formulation<br />

Label claim (mg) Amount found (mg) Label claim %<br />

Sample Ramipril Olmesartan Ramipril Olmesartan Ramipril Olmesartan<br />

1 10 10 10.08 10.18 100.8 101.8<br />

2 10 10 10.11 10.17 101.1 101.7<br />

3 10 10 9.99 10.11 99.9 101.1<br />

4 10 10 10.04 10.18 100.4 101.8<br />

5 10 10 9.98 9.88 99.8 98.8<br />

Mean 10.04 10.10 100.4 101.0<br />

S.D. 0.0561 0.1285 0.561 1.285<br />

Figure 2: Calibration graph of Ramipril at 240 nm.<br />

Figure 3: Calibration graph of Olmesartan at 246 nm.<br />

Figure 4: First derivative spectra of Ramipril, Olmesartan, Synthetic mixture and Tablet formulation.<br />

80 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Santosh R. Karajgi et al.: Simultaneous First Derivative UV Spectrophotometric Estimation of Ramipril and Olmesartan<br />

to 10 μg/ml with methanol, weighed quantities of bulk<br />

drugs were added at four levels, i.e. 50 %, 75 %, 100 %<br />

and 125 % for both drugs and the total drug contents<br />

were determined as described for formulation. The<br />

percentage recovery was determined (Table 2).<br />

RESULTS AND DISCUSSION<br />

Simultaneous analysis of Ramipril and Olmesartan by zero<br />

order UV-Visible spectrophotometric method appears to<br />

be quite impossible because of the total overlap of bands.<br />

(Figure 5). But derivative spectrophotometric technique<br />

involves the differentiation of the normal spectrum with<br />

respect to the wavelength. The fi rst derivative spectra of<br />

the Olmesartan shows an absorbance at 246 nm, where<br />

the absorption for Ramipril is zero and Olmesartan can<br />

be specifi cally measured at that wavelength. Ramipril<br />

on the other hand, has absorbance at 240 nm where<br />

absorbance for Olmesartan is zero; hence Ramipril is<br />

specifi cally measured at that wavelength.<br />

Figure 5: Overtain zero order spectra of Ramipril and Olmesartan.<br />

Table 2: Recovery study of marketed formulation<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 81<br />

Drug<br />

Amount present in<br />

formulation (μg/ml)<br />

Amount<br />

added (%)<br />

Mean<br />

Recovery (%)<br />

Ramipril 10.11 50 99.62<br />

75 100.93<br />

100 98.89<br />

125 99.85<br />

Olmesartan 10.04 50 100.43<br />

75 99.93<br />

100 100.18<br />

125 99.75<br />

The percentage RSD values in precision shows that<br />

proposed method provides acceptable variation of<br />

Ramipril and Olmesartan which were found to be less<br />

than 2% shows its capacity to remain unaffected by<br />

small variations in method parameters and provides<br />

an indication of its reliability during normal usage.<br />

The good recovery values indicate the accuracy of the<br />

Table 3: Validation of the proposed method<br />

Sl. No. Parameters<br />

Ramipril<br />

Experimental value<br />

Olmesartan<br />

ICH Limits<br />

1 Beer’s Range 5–50 μg/ml 5–50 μg/ml –<br />

2 Linearity (R2 ) 0.9994 0.9996 0.9990<br />

3 Precision (% RSD) 0.1754 0.0349 2.0000<br />

4 Accuracy (% Recovery) 99.62–100.93 % 99.75–100.43 % 97–103 %<br />

5 Intercept -0.0001 +0.0078 –<br />

6 Slope +0.0010 +0.0364 –


Santosh R. Karajgi et al.: Simultaneous First Derivative UV Spectrophotometric Estimation of Ramipril and Olmesartan<br />

method (Table 3). Method validation was carried out as<br />

per ICH guidelines. 27<br />

CONCLUSION<br />

The described method gives accurate and precise results<br />

for determination of Ramipril and Olmesartan mixtures<br />

in tablets without prior separation and are easily applied<br />

for routine analysis. This method also provides simple<br />

and reproducible quantitative analysis without any<br />

interference from the excipients.<br />

REFERENCES<br />

1. Macdougall I.C. The role of ACE inhibitors and angiotensin II receptor<br />

blockers in the response to epoetin. Nephrol. Dial. Transplant<br />

1999;14(8):1836–41.<br />

2. Mohamed Saleem T.S., Bharani K., Gauthaman K. Angiotensin II<br />

receptor antagonists: A useful combination therapy for ischemic heart<br />

disease. Emergency Medicine 2010;2:51–9.<br />

3. Hassan Y., Aboul enein, Thiffault C. Determination of Ramipril<br />

and Its Precursors by Reverse Phase High Performance Liquid<br />

Chromatography. Analytical Letters 1991;24(12):2217–24.<br />

4. Al-Majed A.A., Belal F., Abadi A., et al. The voltammetric study and<br />

determination of Ramipril in dosage forms and biological fl uids. Farmaco<br />

2000;55(3):233–8.<br />

5. Zhu Z., Vachareau A., Neirinck L. Liquid chromatography–mass<br />

spectrometry method for determination of Ramipril and its<br />

active metabolite Ramiprilat in human plasma J. Chromatogr. B<br />

2002;779(2):297–306.<br />

6. Ayad M.M., Abdalla A.S., Hisham E., et al. Spectrophotometric and<br />

AAS determination of Ramipril and enalapril through ternary complex<br />

formation. J. Pharm. Biomed. Anal. 2002;28(2):311–21.<br />

7. Rahman N., Ahmad Y., Azmi S.N.H. Kinetic Spectrophotometric Method<br />

for the Determination of Ramipril in Pharmaceutical Formulations.<br />

AAPS Pharm. Sci. Tech. 2005;6(3): E543-E551.<br />

8. Baing M.M., Vaidya V.V., Sane R.T., et al. Simultaneous RP-LC<br />

Determination of Losartan Potassium, Ramipril, and Hydrochlorothiazide<br />

in Pharmaceutical Preparations. Chromatographia 2006;64:293–6.<br />

9. Gowda K.V., Mandal U., Selvan P.S., et al. Liquid chromatography<br />

tandem mass spectrometry method for simultaneous determination of<br />

metoprolol tartrate and Ramipril in human plasma. J. Chromatogr. B.<br />

2007;858:13–21.<br />

10. Patel C.V., Khandhar A.P., Captain A.D., et al. Validated absorption<br />

factor spectrophotometric and Reversed phase High performance<br />

liquid chromatographic methods for the determination of Ramipril and<br />

Olmesartan medoxomil in pharmaceutical formulations. Eurasian J.<br />

Anal. Chem. 2007;2(3):159–71.<br />

11. Liu D., Hu P., Matsushima N., et al. Quantitative determination of<br />

Olmesartan in human plasma and urine by liquid chromatography<br />

coupled to tandem mass spectrometry. J.Chromatogr. 2007;856<br />

(1–2):190–7.<br />

12. Shah N.J., Suhagia B.N., Shah R.R., et al. Development and validation<br />

of a simultaneous HPTLC method for the estimation of Olmesartan<br />

Medoxomil and hydrochlorothiazide in tablet dosage form. Ind. J.<br />

Pharm. Sci. 2007;69(6):834–6.<br />

13. Lincy J., George M., Ranga Rao V.B. Simultaneous estimation of<br />

atorvastatin & Ramipril by RP-HPLC & Spectroscopy. J. Pharm. Sci.<br />

2008;21(3):282–4.<br />

14. Yuan B., Wang X., Zhang F., et al. Simultaneous Determination of<br />

Ramipril and Its Active Metabolite Ramiprilat in Human Plasma by LC–<br />

MS–MS. Chromatographia 2008;68:533–9.<br />

15. Kadukara S.S., Ranjanea P.N., Ranhera S.S., et al. Spectrophotometric<br />

Methods for Determination of Olmesartan Medoxomil and<br />

Hydrochlorothiazide in Tablet Dosage Form. The Pharma Review<br />

2008.<br />

16. Garg G., Saraf S., Saraf S.S. Simultaneous estimation of atorvastatin<br />

& Ramipril by RP-HPLC & Spectroscopy. J. Ind. Chem. Soc.<br />

2008;85(7):769–72.<br />

17. Thamake S.L., Jadhav S.D., Pishawikar S.A. Development and<br />

Validation of Method for Simultaneous Estimation of Atorvastatin<br />

Calcium and Ramipril from Capsule Dosage Form by First Order<br />

Derivative Spectroscopy Asian J. Res. Chem. 2009;2(1):52–3.<br />

18. Patil P.R., Rakesh S.U., Dhabale P.N., et al. Simultaneous Estimation of<br />

Ramipril and Amlodipine by UV Spectrophotometric Method. Reseach.<br />

J. Pharm. and Tech 2009;2 (2):304–7.<br />

19. Karajgi S.R., Simpi C.C., Zambare Y.B. Simultaneous Estimation<br />

of Atorvastatin and Ramipril by First Derivative Spectrophotometric<br />

method. J. Pharm. Res. 2009;2(5):874–7.<br />

20. Kurade K. B., Pai V.P., Gude M.G. RP-HPLC estimation of Ramipril and<br />

telmisartan in tablets. Ind. J. Pharm. Sci. 2009;71(2):148–51.<br />

21. Gupta Y. isocratic RP-HPLC method development & validation for the<br />

simultaneous estimation of Ramipril and Telmisartan in tablet dosage<br />

form. Asian J. Pharm. Clin. Res. 2009;2(4):2441.<br />

22. Bankey S., Tapadiya G.G., Saboo S.S., et al. Simultaneous<br />

Determination of Ramipril, Hydrochlorothizide and Telmisartan by<br />

Spectrophotometry. Int. J. ChemTech Res. 2009;2:183–8.<br />

23. Panchal H.J., Suhagia B.N., Patel N.J., et al. Simultaneous Estimation<br />

of Atorvastatin Calcium, Ramipril and Aspirin in Capsule Dosage Form<br />

by RP-LC. J.Chromatogr. 2009;69:1–2.<br />

24. Qutab S.S., Razzak S.N., Ashfaq M. Simultaneous quantitation of<br />

Olmesartan Medoxomil and Amlodipine besylate in combined Tablets<br />

using HPLC. J. Chili. Chem. Soc. 2009;54 (3):234–7.<br />

25. Rote A.R., Bari P.D. Ratio Spectra Derivative and Zero-Crossing<br />

Difference Spectrophotometric Determination of Olmesartan Medoxomil<br />

and Hydrochlorothiazide. AAPS Pharm. Sci. Tech. 2009;10(4):1200.<br />

26. Wankhede S.B., Wadkar S.B., Raka K.C., et al. Simultaneous estimation<br />

of amlodipine besilate and Olmesartan Medoxomil in pharmaceutical<br />

dosage form. Ind. J. Pharm. Sci. 2009;71 (5):563–7.<br />

27. ICH Harmonized Tripartite Guideline. Text on Validation of Analytical<br />

Procedures. Q2A and Q2B 1995:1–5 and 1–7.<br />

82 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


Purifi cation and characterization of thermostable<br />

amylase from a strain of thermoactinomyces<br />

thalpophilus KSV 17<br />

K. Sreenivasa Rao 1 , P. Ellaiah 2 , Karnakumar V. Biradar 3<br />

1 Department of Pharmaceutics, RRKS College of Pharmacy, Bidar-585402, India<br />

2 Department of Biotechnology, College of Pharmaceutical Sciences, Visakhapatnam, Andhra University, Andra Pradesh, India<br />

3 Department of Pharmacology, RRKS College of Pharmacy, Bidar-585402, India<br />

ABSTRACT<br />

This research reported the Purifi cation and Characterization of Thermostable Amylase from a strain of<br />

T. Thalpophilus KSV 17. The result showed that the purifi ed enzyme specifi c activity of 145.80 U mg –1 , this was<br />

an increase of 21 fold than the crude enzyme extract. The analysis of SDS- polyacrylamide gel electrophoresis<br />

showed that the molecular weight of the enzyme was 52 kDa. The Optimum pH of the purifi ed enzyme showed<br />

maximum activity at pH 7.0, but the enzyme was stable in the pH range of 5.5–7.0. The optimum temperature<br />

of the purifi ed enzyme was 85°C in absence of 10 mM CaCl while 90°C in presence of 10 mM CaCl K and<br />

2 2., m<br />

V values for the purifi ed enzyme were calculated as 5.2 mg ml max –1 , 0.45 mg ml –1 /minute respectively. The thermal<br />

stability of the purifi ed enzyme at 80°C in absence of CaCl and 85°C in presence of CaCl . The purifi ed enzyme<br />

2. 2<br />

mostly inhibited by diethyl pyrocarbonate and N-bromosuccinimide and at 5 mM conc. Ca2+ , Na + and Mg2+ showed stimulatory effect while Cu2+ , Zn2+ , Hg2+ and Mn2+ shown inhibitory effect. The purifi ed enzyme showed<br />

good stability and compatibility on commercial detergents and on being stored for 60 minutes at 85°C.<br />

Key words: Purifi cation, Characterization, α-Amylase, Sephadex G-200, SDS-PAGE<br />

INTRODUCTION<br />

Microbial enzymes and coenzymes are<br />

widely used in several industries, notably<br />

in detergent, food processing, brewing<br />

and pharmaceuticals. They are also used<br />

for diagnostic, scientifi c and analytical<br />

purposes. Since ancient times they have<br />

been used in the preparation of fermented<br />

foods, especially in oriental countries. 1<br />

The activity of an enzyme is determined<br />

by the enzyme concentration, substrate<br />

concentration and its availability,<br />

concentration of co-factors, allosteric<br />

effectors, the concentration and type of<br />

inhibitors, ionic strength, pH, temperature<br />

and time since start of the reaction. The<br />

way in which each of these parameters<br />

affects enzyme activity can be determined<br />

by studying the enzyme kinetics, many assay<br />

procedures for determining the enzyme’s<br />

activity are available. Some of them are<br />

typically dependent on measurement of<br />

chemical changes Eg: Biuret Method,<br />

Lowry Method, Bradford Method etc.<br />

and others are based on physical changes.<br />

Some of these assays are also designed<br />

to investigate particular properties of the<br />

enzyme preparations and physical changes<br />

Terms, which are frequently encountered,<br />

especially in investigations of enzyme<br />

purifi cation and use are, specifi c activity and<br />

molecular activity per milligram of enzyme.<br />

Where the enzyme preparation is not pure,<br />

specifi c activity is frequently expressed in<br />

units per milligram of protein or in units<br />

per milligram of enzyme preparation. Due<br />

to wide range of application of α-Amylase,<br />

in order to get not only high quality and but<br />

Research Ar cle<br />

Received Date : 26-12-2011<br />

Revised Date : 04-03-2012<br />

Accepted Date : 06-03-2012<br />

DOI: 10.5530/rjps.2012.1.12<br />

Address for<br />

correspondence<br />

K.Sreenivasa Rao<br />

R.RK.S. College of Pharmacy<br />

Naubad, Bidar-585401,<br />

Karnataka, India<br />

Email: bidarkaran@gmail.com<br />

www.rjps.in<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 83


K. Sreenivasa Rao et al.: Purifi cation and Characterization of Thermostable Amylase from a Strain of Thermoactinomyces thalpophilus KSV 17<br />

also high grade α-Amylase we made objectives behind<br />

the sudy.<br />

In this research, α-amylase was purifi ed from a strain of<br />

T. Thalpophilus KSV 17 in three steps of purifi cation<br />

processes, namely precipitation with ammonium sulfate,<br />

precipitation with zink sulfate and Sephadex G-200<br />

gel fi ltration chromatography. The enzyme was then<br />

characterized. The enzyme activity was determined<br />

based on Bernifi eld method, 2 while protein content<br />

was determined by Lowry method. 3<br />

MATERIAL AND METHODS<br />

Chemicals and Microorganism Used<br />

Agarose, acrylamide, bis-acrylamide, sodium dodecyl<br />

sulfate (SDS), TEMED, ammonium per sulfate and<br />

other chemicals for polyacrylamide gel electrophoresis<br />

(SDS-PAGE) were purchased from Sigma Chemical Co.,<br />

U.S.A. A strain of T. thalpophilus mutant KSV 17 was<br />

used for the study. The research phases done were as<br />

follows: Production, Purifi cation and Characterization<br />

of purifi ed enzyme.<br />

Production of α-Amylase<br />

The amylase production was carried out in a 2 liter<br />

fermentor, containing 1.5 L modifi ed production<br />

medium containing Starch 2%, Yeast extract 0.05%, Mg<br />

SO 4 0.01 and salt solution (KH 2 PO 4 0.15%, K 2 HPO 4<br />

0.22% CaCl 2 .2H 2 0 0.0025% and FeSO 4 . 7H 2 0 0.00025%).<br />

A 10% (v/v) level of inoculum was added and the<br />

fermentor was run at 55°C for 48h. Whole fermentation<br />

broth was centrifuged at 10,000 g at 4°C and the clear<br />

supernatant was separated. The supernatant (crude<br />

enzyme) was subjected to recovery and purifi cation<br />

processes.<br />

Purifi cation of α-Amylase<br />

The purifi cation of enzyme was done by following steps,<br />

Ammonium Sulfate Precipitation<br />

Ammonium sulfate was added at different<br />

concentrations ranging from 40 to 80 % saturation.<br />

The precipitates so obtained were suspended in cold<br />

saline solution (2ml) and tested for amylase activity and<br />

total protein content. The salting out concentration of<br />

crude enzyme was established to be 60% on the basis<br />

of enzyme activity. To obtain complete precipitation<br />

of the crude enzyme, the remaining harvest fl uid was<br />

subjected to ammonium sulfate precipitation at 60%<br />

saturation. For this purpose, solid ammonium sulfate<br />

(195g) was added gradually with mechanical stirring<br />

to 500ml of harvest fl uid at 4˚C to a saturation of<br />

60%. The precipitate so formed was separated by<br />

centrifugation (8000g) for 15 min., again suspended in<br />

cold saline solution (100ml) and dialyzed in cold against<br />

1 L of 0.05M Phosphate buffer (pH 7) for 20 h. After<br />

dialysis, the solution was centrifuged and supernatant<br />

obtained was designated as fraction-I. It was used for<br />

zinc sulfate precipitation.<br />

Zinc Sulfate Precipitation<br />

About 100ml of the pooled supernatant (fraction-I)<br />

with activity of 20 U/mg protein was diluted with<br />

1.4 L of 0.05M Phosphate buffer (pH 7) to obtain 2<br />

units of absorbance at 280 nm. To this solution 75 ml<br />

of 0.1M zinc sulfate was added drop wise with stirring<br />

at 0˚C in an ice bath. The precipitate thus formed was<br />

separated by centrifugation (8000g) for 15 min. and<br />

re-suspended in 50ml of 0.4M sodium citrate. This<br />

solution was dialyzed in cold (4ºC) against 240 ml of<br />

0.05M Phosphate buffer, pH 7. The resulting solution<br />

was designated as fraction-II, and subjected to gel<br />

fi ltration chromatography.<br />

Sephadex G-200 Gel Filtration Chromatography<br />

The dialyzed enzyme (fraction-II) was centrifuged at<br />

8000rpm for 15 min. and supernatant was chromatographed<br />

on a column of Sephadex G-200. The sample<br />

(fraction-II) was loaded onto a column of Sephadex<br />

G-200 (1.5cm × 24cm) equilibrated with 0.05M phosphate<br />

buffer (pH 7). The column was eluted at a fl ow<br />

rate of 4.0ml /h with a discontinuous gradient from<br />

0.1 M to 0.8 M phosphte buffrer in the same buffer.<br />

A total of 25 fractions were collected. Fractions (15–18)<br />

with high amylase activity were pooled together, dialyzed,<br />

concentrated by lyophilization and used for further<br />

studies. It was labeled as fraction-III.<br />

Sodium Dodecyl Sulfate Polyacrylamide<br />

Gel Electrophoresis (SDS-PAGE)<br />

After Sephadex G-200 column chromatography, the<br />

fractions (19–23) showing the highest specifi c activity<br />

were pooled, dialyzed, lyophilized and then subjected to<br />

SDS-PAGE. The SDS-PAGE was performed according<br />

to Laemmli (1970) 4 using 12% acrylamide.<br />

Characterization of Purifi ed Enzyme<br />

The Characterization of purifi ed enzyme includes:<br />

Determination of optimum pH, Temperature, effects<br />

of metal ions, effects of inhibitors and chelators and<br />

stability towards temperature, pH, and detergents, kinetic<br />

data, Activity test of α-amylase and protein content.<br />

Determination of Optimum pH of Purifi ed Enzyme<br />

The pH was adjusted using the following buffers: 0.05 M<br />

phosphate (pH 5.0–7.0), Tris-HCl (pH 8.0) and glycine-<br />

NaOH (pH 9.0–12.0). Reaction mixtures were incubated<br />

at 55ºC for 30 min. and the activity of the enzyme<br />

84 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


K. Sreenivasa Rao et al.: Purifi cation and Characterization of Thermostable Amylase from a Strain of Thermoactinomyces thalpophilus KSV 17<br />

was measured. For stability study, the purifi ed enzyme<br />

was diluted in different relevant buffers (pH 6.0–12.0)<br />

in two sets. One set incubated for 2h and another set for<br />

20 h at 55ºC. The relative activity at each exposure was<br />

measured as per assay procedure.<br />

Determination of Optimum Temperature<br />

of Purifi ed Enzyme<br />

The activity of the purifi ed enzyme was determined by<br />

incubating the reaction mixture at different temperatures<br />

ranging from 30 to 95°C for 30 min. in the absence and<br />

presence of 10 mM CaCl 2 . Thermostability of the purifi ed<br />

enzyme was determined in the presence and absence<br />

of CaCl 2 (10mM) by pre-incubating enzyme samples in<br />

phosphate buffer for 30 minutes at temperatures ranging<br />

from 30 to 95ºC.<br />

Determination of Amylase Activity on<br />

Various Metal Ions<br />

Metal ions viz. Ca2+ , Mg2+ , Hg2+ , Fe3+ , Na + , Zn2+ , Mn2+ and Cu2+ (10 mM) were investigated by adding them<br />

into the reaction mixture. The mixture was incubated<br />

for 30 min. at 55°C and the relative amylase activities<br />

were measured.<br />

Determination of Amylase Inhibitors and<br />

Chelators on Purifi ed Enzyme<br />

The effect of various amylase inhibitors (at 5mM), such<br />

as Diethyl pyrocarbonate (DEPC), N-bromo succinimide<br />

(N-BS) cysteine-inhibitors p-chloromercuric benzoate<br />

(pCMB) and β-mercaptoethanol (β-MCE), and a<br />

chelator of divalent cations [Ethylene diamine tetra<br />

acetic acid (EDTA)] were determined by preincubation<br />

with the enzyme solution for 30 min at 55°C before the<br />

addition of substrate. The relative amylase activity was<br />

measured.<br />

Determination of Enzyme Stability in<br />

Presence of Detergents<br />

The compatibility of KSV-17 amylase with local<br />

laundry detergents was investigated in the presence of<br />

10 mM CaCl . The detergents were diluted in distilled<br />

2<br />

water (0.7% w/v), incubated with 0.1ml of enzyme<br />

(500U/ml) for 4 h at 55°C and the residual activity was<br />

determined.<br />

Determination of Enzyme Kinetic Data<br />

of Purifi ed Enzyme<br />

Initial rates of soluble starch hydrolysis was determined<br />

with various concentrations at various substrate<br />

concentrations ( g/100ml: 0.10, 0.20, 0.30, 0.40, 0.50,<br />

0.75, 1.00, 1.25, 1.50, 1.75 and 2.00). The kinetic<br />

constants K m and V max were determined by the method<br />

of Lineweaver-Burk.<br />

Activity Test of α-Amylase and Protein<br />

Content Determination<br />

The α- amylase activity was done using 3, 5-dinitro<br />

salicylic acid as a coupling agent by following mandels<br />

(1976) method. The content of protein was determined<br />

based on method of lowry et al. (1951). 3<br />

RESULTS AND DISCUSSION<br />

Table 1: Summary of purifi cation steps of amylase from T. Thalpophilus KSV 17<br />

Purifi cation step<br />

Total enzyme<br />

activity (U)<br />

Total<br />

protein (mg)<br />

Purifi cation of Amylase of Thermoactinomyces<br />

Thalpophilus KSV 17<br />

The enzyme production was carried in a 2 L fermentor<br />

and the extraction of growth media that contain the<br />

extracellular enzyme with cold centrifugation to get<br />

the raw extract enzyme which further precipitated<br />

by salt of ammonium sulfate at 60% of saturation<br />

grade, and obtained supernatant precipitated with zinc<br />

sulfate, resulting solution was subjected for gel fi ltration<br />

column chromatography using Sephadex G-200, then<br />

subjected for SDS-PAGE was performed according<br />

to Laemmli et al.(1970) 4 using 12% acrylamide fi naly<br />

α- amylase activity was done using 3,5-dinitro salicyclic<br />

acid as a coupling agent according to Mande l (1976)<br />

method. Purifi cation steps were showed in Table1.<br />

Gel fi ltration Chromatography using<br />

Sephadex G-200<br />

The purifi cation of enzyme with Gel fi ltration<br />

chromatography using Sephadex G-200, used phosphate<br />

buffer 0.05M, pH 7 and column eluted with 0.1 M to<br />

0.8 M NaCl as elution buffer. The protein pattern (A ) 280<br />

and activity of α-amylase obtained from gel fi ltration<br />

chromatography using Sephadex G-200. From the<br />

fi gure 1 showed only one peak showed α-amylase activity<br />

observed at 0.4M NaCl concentration that was fraction<br />

(15th–18th), the highest activity was fraction 18th as<br />

showed in fi gure 1.<br />

Specifi c<br />

activity (U/mg)<br />

Purifi cation<br />

fold % Recovery<br />

Crude enzyme 175000 26800 6.52 1.0 100<br />

(NH4)2SO4 precipitation 131000 7890 16.60 2.5 74.85<br />

Sephadex G-200 13560 93 145.80 22.3 7.74<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 85


K. Sreenivasa Rao et al.: Purifi cation and Characterization of Thermostable Amylase from a Strain of Thermoactinomyces thalpophilus KSV 17<br />

Figure 1: Chromatography of α-amylase from Thermoactinomyces thalpophilus KSV 17 at Gel filtration column chromatography using<br />

Sephadex G-200, elution solution of buffer phosphate 0.05M, pH 7.0, column size 1.5 × 24 cm and flow rate 1.0 ml/h.<br />

Homogenity test of Purifi ed Enzyme by SDS-<br />

Polyacrylamide gel Electrophoresis<br />

The result of electrophoresis SDS-polyacrylamide of<br />

purifi ed enzyme can be seen in electrophoregram as<br />

shown in Figure 2. This fi gure showed that the molecular<br />

weight of purifi ed enzyme was 52 kDa.<br />

Characterization of Purifi ed Enzyme<br />

Determination of Optimum pH of Purifi ed Enzyme<br />

The activity (%) of purifi ed enzyme and stability can<br />

be seen in fi gure 3. It showed optimum pH of purifi ed<br />

enzyme is 6.0 and stable in the pH range of 4–7.5 in<br />

case of 2h. preincubation and at 20h. preincubation the<br />

enzyme was stable in the pH range 5.5–7.0 (Figure 3).<br />

Determination of Optimum Temperature<br />

of Purifi ed Enzyme<br />

The activity (%) of purifi ed enzyme at various<br />

temperatures in presence and absence of CaCl 2 can be<br />

seen in fi gure 4. The recorded optimum temp of purifi ed<br />

enzyme was 85ºC in the absence of CaCl 2 , while 90ºC in<br />

the presence of CaCl 2 (Figure 4).<br />

Determination of Inhibitors and Chelators<br />

effect on Purifi ed Enzyme<br />

The inhibitors tested (at 5 mM conc.), diethyl<br />

pyrocarbonate (DEPC) was able to inhibit the almost<br />

completely, N-bromo succinimide (N-BS) exhibited<br />

89% inhibition. Results are similar to Bolton et al.,<br />

(1997) 5 study. This indicated that it is an alpha amylase<br />

and, 51% inhibition was observed with EDTA and slight<br />

stimulation effect by β -mercaptoethanol (fi gure 5).<br />

Determination of Compatibility with<br />

Detergents on Purifi ed Enzyme:<br />

Besides pH, a good detergent amylase is expected to<br />

be stable in presence of commercial detergents. The<br />

amylase from KSV 17 showed excellent stability and<br />

compatibility in the presence of locally available detergents<br />

(Nirma, Wheel, Henko, Surf excel, Ariel and Rin).<br />

Determination of Metal ions Effect on<br />

Purifi ed Enzyme<br />

The data indicates that Ca 2+ , Mg 2+ and Na + have slight<br />

stimulatory effect activation of the enzyme may be<br />

due to activation of metal ions while other ions have<br />

slight inhibitory effect on the enzyme. These results<br />

suggest, the metal ions apparently protected the enzyme<br />

against thermal denaturation and played a vital role in<br />

maintaining the active confi rmation of the enzyme at high<br />

temperatures (Donaghy and McKay (1993) 6 (Figure 6).<br />

Determination of Kinetic data of<br />

Purifi ed Enzyme<br />

The graph of Determination of K and V values<br />

m max<br />

of the purifi ed enzyme can be seen in fi gure 7. From<br />

the graph, the K (Michaelis constant) and V m max<br />

were calculated as 5.2 mg/ml and 0.45 mg/ml/min<br />

respectively. (Figure7). Aguilar et al., (2000) reported<br />

a K of 3.44 mg/ml and V of 0.45 mg/ml/min for<br />

m max<br />

alpha amylase from Lactobacillus. A K of 0.90 g/l was<br />

m<br />

reported for a thertmostable alpha amylase from Bacillus<br />

licheniformis by Ivanova et al., (1993). 7 Krishnan and<br />

Chandra, (1983) 8 reported a K of 1.274 mg/ml and<br />

m<br />

V of 0.738 mg/ml/min for alpha amylase obtained<br />

max<br />

from Bacillus licheniformis CUMC 305.<br />

86 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


K. Sreenivasa Rao et al.: Purifi cation and Characterization of Thermostable Amylase from a Strain of Thermoactinomyces thalpophilus KSV 17<br />

Figure 2: SDS-PAGE of Amylase from T. Thalpophilus KSV 17. Lane A, crude enzyme; Lane B, purified enzyme; Lane C, molecular mass<br />

markers. The molecular weight of standard protein markers used are: BSA (67 kDa), Ovalbumin (45 kDa), Carbonic anhydrase (30 kDa),<br />

Trypsinogen (24 kDa) and α-lactalbumin (14 kDa).<br />

Figure 3: The optimum pH of purified enzyme 6.0, same figure also showed that the purified enzyme was stable in the pH range of 4–7.5<br />

in case of 2h. preincubation group. But in the case of 20 h. preincubation group, the enzyme was stable between pH 5.5 and pH 7.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 87


K. Sreenivasa Rao et al.: Purifi cation and Characterization of Thermostable Amylase from a Strain of Thermoactinomyces thalpophilus KSV 17<br />

Figure 4: For the study temperature ranging from 30 to 90 o C in the absence and presence of 10 mM CaCl 2 . The result indicates purified<br />

enzyme is stable at 80 o C and showed high activity at 85 o C. The optimum temperature recorded was 85 o C for purified enzyme in the<br />

absence of CaCl 2 , while 90 o C in the presence of CaCl 2 .<br />

Figure 5:: The inhibitors like DEPC, N-BS exhibited 89% inhibition., 51% inhibition was observed with EDTA and slight stimulation effect<br />

by α-mercaptoethanol.<br />

CONCLUSION<br />

Based on above results and discussion, the following<br />

conclusions are made: The analysis of SDS- PAGE<br />

indicates the molecular weight of α-Amylase is 52 kDa<br />

depending on the relative mobility. The purifi ed enzyme<br />

specifi c activity was an increase of 21 fold than the crude<br />

enzyme extract. The purifi ed enzyme has optimum<br />

pH 7.0. The enzyme was stable between pH 6 and<br />

88 RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012


K. Sreenivasa Rao et al.: Purifi cation and Characterization of Thermostable Amylase from a Strain of Thermoactinomyces thalpophilus KSV 17<br />

Figure 6: The metal ions Ca 2+ , Mg 2+ and Na + have slight stimulatory effect while other slight inhibitory effect on the enzyme.<br />

Figure 7: Amylase activity done according to Bernfield (1955), using 3,5- dinitro salicylic acid as a coupling agent.<br />

pH 7 even after 20 h. preincubation. The optimum<br />

temperature for purifi ed enzyme activity was 85°C<br />

in absence of CaCl 2 while 90ºC in the presence of<br />

CaCl 2 . Amylase inhibitors like DEPC, N-BS and<br />

pCMB showed exhibitory however Ca 2+ , Mg 2+ and<br />

Na + ions have slight stimulatory effect. This purifi ed<br />

enzyme showed good compatibility and stability with<br />

commercial detergents.<br />

REFERENCES<br />

1. Reed G. Enzymes in Food Processing, 2nd ed, Academic Press,<br />

Orlando, 1975, 256–60.<br />

2. Bernfeld P. Amylases, α and β, Methods in enzyme, Vol. I, Academic<br />

Press, New York, 1955, 149–58.<br />

3. Lowry OH. Rosebrough N J, Farr AL, Randall RJ. Adenyl cyclase<br />

activity in human platelets. J Biol Chem1951;193:265–71.<br />

4. Laemmli UK. Purifi cation and Biochemical Charectirisation of Ricin fom<br />

Castor. Nature1970;227:680–7.<br />

5. Bolton DJ, Kelly CT, Fogarty WM. Purifi cation and charecterization of<br />

Amylase of Bacillus. Enz Microb Technol 1997;20:340–3.<br />

6. Ivanova VN, Dobreva EP, Emanuilova EI. Efect of temparature on some<br />

charecteristics thermostable Amylase enzyme. J Biotechnol 1993;28:<br />

277–89.<br />

7. Krishnan T, Chandra AK. Effect of oilseed cakes on alpha-amylase<br />

production by Bacillus licheniformis CUMC305. Appl Environ Microbiol<br />

1982;44:270–4.<br />

8. Domingues CM, Peralta RM. Production of amylase by soil fungi and<br />

partial biochemical characterization of amylase of a selected strain<br />

(Aspergillus fumigatus Fresenius). Can. J Microbiol 1993;39:681–5.<br />

RGUHS J Pharm Sci | Vol 2 | Issue 1 | Jan–Mar, 2012 89

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!