18.03.2015 Views

26 July 2009 Cardiology and Oncology Drug ... - ICON plc

26 July 2009 Cardiology and Oncology Drug ... - ICON plc

26 July 2009 Cardiology and Oncology Drug ... - ICON plc

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.

March <strong>2009</strong> Vol 9 No 3<br />

www.drugdeliverytech.com<br />

IN THIS<br />

ISSUE<br />

INTERVIEW WITH<br />

DEPOMED’S<br />

PRESIDENT & CEO<br />

CARL A. PELZEL<br />

Combination<br />

Devices 18<br />

Abhijit Gokhale, PhD<br />

Excipient<br />

Sourcing 22<br />

Alen Guy, PhD<br />

Solid Dose<br />

Injection 24<br />

Charles Potter, PhD<br />

Nanomedicine<br />

Market 32<br />

Bill Martineau, MBA<br />

FEATURING<br />

The science & business of drug development in specialty pharma, biotechnology, <strong>and</strong> drug delivery<br />

Xiaochun Yu,<br />

PhD<br />

A New Approach to<br />

Threshold Evaluation &<br />

Quantitation of<br />

Unknown Extractables<br />

& Leachables Using<br />

HPLC/CAD<br />

Ms. Cindy H.<br />

Dubin<br />

Proteins & Peptides:<br />

Dependent On<br />

Advances in <strong>Drug</strong><br />

Delivery?<br />

Mr. Mike<br />

Mesa<br />

From Battlefield to<br />

Backpack: Evolution<br />

of the Auto-Injector<br />

Analytical<br />

Testing Labs 62<br />

<strong>Cardiology</strong> &<br />

<strong>Oncology</strong> 68<br />

Stuart L. Cantor, PhD


Therapeutic<br />

Focus<br />

<strong>Cardiology</strong> & <strong>Oncology</strong> <strong>Drug</strong> Development<br />

& Regulation<br />

SPECIALTY PHARMA MARCH <strong>2009</strong> Vol 9 No 3<br />

68<br />

By: Stuart L. Cantor, PhD, Senior Scientist, <strong>and</strong> Kadriye Ciftci, PhD, Senior Director <strong>Drug</strong> Delivery,<br />

<strong>ICON</strong> Development Solutions<br />

Introduction<br />

Heart disease <strong>and</strong> cancer are still the<br />

two leading causes of mortality in the<br />

world. Recent data show that in the US,<br />

the total direct <strong>and</strong> indirect costs<br />

associated with treating cardiovascular<br />

diseases <strong>and</strong> stroke are estimated to be<br />

$449 billion, by comparison, the<br />

estimated costs for all types of cancer<br />

was $219 billion. 1 Sales of drugs treating<br />

cardiovascular disorders, hypertension,<br />

<strong>and</strong> cancer accounted for roughly 31% of<br />

the $287 billion prescription<br />

pharmaceutical market in 2007; these<br />

drugs continue to be the largest therapy<br />

classes in the US (Figure 1). 2 In the<br />

cardiology sector, Pfizer’s Lipitor ®<br />

amassed sales revenues of $12.7 billion<br />

in 2007, making it the best selling drug<br />

in pharmaceutical history. Not to be<br />

outdone, the biotech industry has<br />

likewise developed a number of<br />

successful blockbuster cancer therapies<br />

as intravenous solutions or vaccines,<br />

including Herceptin ® <strong>and</strong> Avastin ® ,<br />

monoclonal antibody-based therapies to<br />

treat breast <strong>and</strong> colon cancer,<br />

respectively, <strong>and</strong> Gardasil ® , a vaccine<br />

against cervical cancer. 3 Sales of cancer<br />

drugs are projected to double <strong>and</strong> could<br />

reach $80 billion by 2012, according to<br />

IMS Health.<br />

There are several reasons for this<br />

increased sales growth, which include<br />

extended lifespan, increases in obesity<br />

<strong>and</strong> hypertension in the US population,<br />

an increasing number of patients on<br />

chemotherapy in major markets, longer<br />

treatment periods for a growing number<br />

of patients, <strong>and</strong> the<br />

greater availability<br />

of more expensive<br />

<strong>and</strong> modern<br />

targeted therapies<br />

to treat these<br />

diseases. However,<br />

hotly debated<br />

issues have risen<br />

over the long-term<br />

safety of coronary<br />

drug-eluting stents<br />

(DES), efficacy of<br />

beta-blockers in<br />

treating<br />

hypertension, <strong>and</strong><br />

the long-term<br />

safety of statins. In<br />

the oncology<br />

sector, there has<br />

Figure 1.<br />

been an increase in<br />

the development of highly potent,<br />

hydrophobic compounds, <strong>and</strong> difficulty<br />

in ensuring their solubility as well as<br />

specificity to target the tumor site.<br />

Because oncology drugs are cytotoxic,<br />

maintaining containment facilities during<br />

the developmental phase for these actives<br />

can become expensive. Furthermore,<br />

with both of these drug classes,


difficulties proving efficacy or<br />

unexpected safety concerns, particularly<br />

during later-stage development amid<br />

Phase III trials, can be particularly<br />

challenging.<br />

The increased number of<br />

blockbuster drugs that are scheduled to<br />

lose patent protection in the coming<br />

years (Figure 2), coupled with the FDA’s<br />

Amendments Act of 2007 granting<br />

sweeping new powers to the agency for<br />

such tasks as requiring drug makers to<br />

do post-marketing clinical trials, are<br />

making the regulatory climate more<br />

expensive <strong>and</strong> time-consuming for the<br />

pharmaceutical industry. Furthermore,<br />

because drug safety issues are<br />

paramount due to highly publicized<br />

cases like Vioxx®, the FDA is closely<br />

scrutinizing safety data while improving<br />

its management of safety signals.<br />

<strong>Drug</strong> Development<br />

<strong>Oncology</strong> drugs in development are<br />

generally highly potent as well as<br />

cytotoxic; some may even have narrow<br />

therapeutic windows. It is important to<br />

assess the safety of these compounds<br />

early on to determine that they are<br />

effective in treating the disease process<br />

while not damaging healthy tissue.<br />

According to the M3 Guidance for<br />

Industry, single-dose acute toxicity<br />

studies are required to be performed for<br />

pharmaceuticals <strong>and</strong> should be<br />

evaluated in two small mammalian<br />

model species before the first human<br />

exposure. 4<br />

The ratio of time for animal to<br />

human testing is 1:1, meaning that the<br />

FDA will allow a company to conduct<br />

human clinical trials for only the same<br />

time period in which animal data has<br />

been supplied. Sometimes, the agency<br />

can also request that safety <strong>and</strong>/or<br />

toxicity data in a non-human primate<br />

also be supplied before an<br />

Investigational New <strong>Drug</strong> (IND) can be<br />

approved. A contract research<br />

organization<br />

(CRO) can assist a<br />

company with<br />

developing a<br />

game plan for the<br />

extent of both<br />

preclinical animal<br />

testing <strong>and</strong> future<br />

clinical trials;<br />

such services can<br />

be outsourced by<br />

start-up or virtual<br />

companies with<br />

limited in-house<br />

drug development<br />

resources. The<br />

CRO can also<br />

help the company<br />

in its<br />

correspondence<br />

Table 2.<br />

with the FDA <strong>and</strong><br />

offer guidance<br />

during the critical pre-IND meeting.<br />

Under current regulations, a sponsor is<br />

permitted to start their clinical trials 30<br />

days after filing its IND with the FDA<br />

unless notified earlier that there are<br />

issues with the application.<br />

Recent reports from the<br />

Pharmaceutical Research <strong>and</strong><br />

Manufacturers of America (PhRMA)<br />

noted that 750 new medicines are being<br />

tested in the fight against cancer, <strong>and</strong><br />

256 new medicines are in development<br />

to treat cardiovascular diseases. 5,6 While<br />

there have been a number of successful<br />

anti-neoplastic small molecules<br />

launched over the years, such as 5-<br />

fluorouracil, paclitaxel, <strong>and</strong><br />

doxorubicin, the focus has shifted away<br />

from broad-acting cytotoxic drugs<br />

toward the development of new<br />

therapies directed against specific<br />

molecular targets. Although biologics<br />

<strong>and</strong> vaccines derived from larger protein<br />

molecules offer promise in these<br />

therapeutic areas due to their high<br />

disease specificity <strong>and</strong> activity at<br />

relatively low concentrations as<br />

compared with small molecules,<br />

physicochemical stability <strong>and</strong> potential<br />

immunologic issues need to be closely<br />

monitored <strong>and</strong> controlled to ensure a<br />

drug product’s beneficial effects.<br />

“In contrast to most drugs that are<br />

chemically synthesized <strong>and</strong> with a<br />

known structure, most biologics are<br />

complex mixtures that are not easily<br />

identified or characterized. Biological<br />

products, including those manufactured<br />

by biotechnology, tend to be heat<br />

sensitive <strong>and</strong> susceptible to microbial<br />

contamination,” says Paul Richards,<br />

FDA Spokesperson at CBER.<br />

“Therefore, it is necessary to use aseptic<br />

principles from initial manufacturing<br />

steps in order to ensure sterility of the<br />

finished products, which is also in<br />

contrast to most conventional drugs.”<br />

Furthermore, vaccine clinical<br />

development follows the same general<br />

pathway as for drugs <strong>and</strong> other<br />

biologics. However, due to the complex<br />

nature of many vaccines, each lot must<br />

be thoroughly tested for safety, sterility,<br />

<strong>and</strong> potency by the manufacturer. These<br />

tests, as well as many others that<br />

manufacturers must perform, are<br />

SPECIALTY PHARMA MARCH <strong>2009</strong> Vol 9 No 3<br />

69


SPECIALTY PHARMA MARCH <strong>2009</strong> Vol 9 No 3<br />

70<br />

specified in their biologic license<br />

applications (BLAs). If the product is<br />

subject to official release by the FDA’s<br />

Center for Biologics Evaluation <strong>and</strong><br />

Research (CBER), the manufacturer<br />

must submit samples of each production<br />

lot to the Agency together with a release<br />

protocol showing a summary of the lot<br />

manufacturing history <strong>and</strong> the results of<br />

all the manufacturer’s tests performed on<br />

that lot, says Mr. Richards.<br />

“The most challenging aspects to<br />

developing these biologic drugs is the<br />

design of efficient <strong>and</strong> robust<br />

fermentation <strong>and</strong> purification processes<br />

<strong>and</strong> the production of a stable<br />

formulation,” says Peter Ihnat, PhD, Sr.<br />

Principal Scientist, Protein Therapeutics,<br />

Bristol-Myers Squibb.<br />

Protein formulations can be<br />

developed using either lyophilized<br />

powders or liquid parenterals; however,<br />

freeze-drying is not as popular an option<br />

nowadays due to the increased cost <strong>and</strong><br />

longer development time required to<br />

produce a successful formulation.<br />

“Typically, the target level for<br />

protein impurities for liquid formulations<br />

is < 5%, <strong>and</strong> these are usually due to<br />

oligomers or aggregates; however, due to<br />

the immunogenic potential of these<br />

compounds, impurity levels are<br />

evaluated on a case-by-case basis.<br />

Moreover, long-term protein stability is<br />

monitored using at least two to three<br />

orthogonal methods in addition to an in<br />

vitro bioassay to assess biological<br />

activity,” says Mr. Ihnat.<br />

Regulatory<br />

Submissions<br />

The CTD or Common Technical<br />

Document was developed to be used for<br />

regulatory submissions <strong>and</strong> finalized as<br />

the M4 Guidance for Industry in 2004. 7,8<br />

This table of contents format is highly<br />

recommended for INDs, New <strong>Drug</strong><br />

Applications (NDAs), <strong>and</strong> Abbreviated<br />

NDAs (ANDAs) from the US <strong>and</strong><br />

required for European <strong>and</strong> Japanese<br />

submissions. Canadian INDs must also<br />

be in CTD format; however, these are<br />

referred to as CTAs or Clinical Trial<br />

Applications for Phases I-III. The CTD<br />

contains several modules with Modules 2<br />

<strong>and</strong> 3 containing critical Chemistry,<br />

Manufacturing, <strong>and</strong> Control (CMC)<br />

information.<br />

While Module 2 deals with both<br />

non-clinical <strong>and</strong> clinical overviews <strong>and</strong><br />

summaries, the quality section or<br />

Module 3 deals only with CMC issues<br />

<strong>and</strong> provides information on the<br />

physicochemical properties <strong>and</strong> control<br />

of the drug substance as well as the<br />

development, manufacture, <strong>and</strong> control<br />

of the finished drug product. Modules 4<br />

<strong>and</strong> 5 address non-clinical <strong>and</strong> clinical<br />

study reports, respectively. Updates to<br />

the IND data following Phase I-II trials<br />

can be provided in information<br />

amendments <strong>and</strong> annual reports;<br />

however, the emphasis should be on<br />

reporting significant changes that can<br />

have a safety-related impact. CMC<br />

development will continue in parallel<br />

with the clinical development during<br />

Phase III studies. 9,10<br />

If the CMC section will be written<br />

for an already approved drug but a new<br />

dosage form, the <strong>Drug</strong> Master File<br />

(DMF) number, if available, can be<br />

referenced for some of the pertinent<br />

manufacturing information for the drug<br />

substance. For all scientific data, the<br />

FDA is particularly concerned that the<br />

experimental study design <strong>and</strong> statistical<br />

analysis be sound <strong>and</strong> free from flaws,<br />

<strong>and</strong> requires a rationale <strong>and</strong> justification<br />

used for final specifications selected as<br />

well as the use of novel excipients <strong>and</strong><br />

any unusual tests performed.<br />

Two other regulatory submission<br />

pathways available are the 505(b)(2) <strong>and</strong><br />

combination product options. The<br />

505(b)(2) route offers companies the<br />

advantage of not having to supply the<br />

safety <strong>and</strong> efficacy data on an already<br />

approved drug product, <strong>and</strong> such filings<br />

can be used to support new <strong>and</strong><br />

innovative drug delivery forms. Data<br />

from published studies can even be<br />

submitted to the FDA. However, the<br />

company would need to provide<br />

additional clinical data necessary to<br />

demonstrate any safety <strong>and</strong> efficacy<br />

differences between the original drug<br />

<strong>and</strong> the 505(b)(2) drug. Some of the<br />

different types of applications covered by<br />

the section 505(b)(2) are:<br />

• change in an active ingredient (ie,<br />

different salt, ester, complex,<br />

chelate, clathrate, racemate, or<br />

enantiomer) for a listed drug<br />

containing the same active<br />

moiety;<br />

• change in dosage form, strength,<br />

formulation, dosing regimen, or<br />

route of administration; <strong>and</strong>/or<br />

• change from a prescription<br />

indication to an over-the-counter<br />

indication.<br />

The important benefits of using the<br />

505(b)(2) filing route are a faster<br />

pathway toward regulatory approval<br />

without giving the sponsor the burden of<br />

supplying duplicate safety <strong>and</strong> efficacy<br />

data on an already existing compound.<br />

However, the FDA does still require that<br />

a sponsor provide additional clinical<br />

data, termed bridging studies, which are<br />

necessary to support any changes or<br />

modifications from the listed drug(s) to<br />

the 505(b)(2) drug(s), <strong>and</strong> these studies<br />

will allow extrapolation of the efficacy<br />

<strong>and</strong> safety data. Furthermore, a<br />

505(b)(2) applicant may qualify for 3 or<br />

5 years of Hatch-Waxman marketing<br />

exclusivity. 11<br />

The other non-traditional route for<br />

regulatory approval deals with<br />

combination products, such as coronary<br />

DES, which are considered a drugdevice<br />

combination product under the


Code of Federal Regulations (CFR) 21<br />

CFR 3.2(e)(1). The Office of<br />

Combination Products (OCP) at the FDA<br />

assigns such products to a lead agency<br />

center, based upon the product’s primary<br />

mode of action. OCP ensures timely,<br />

consistent pre-market review <strong>and</strong><br />

appropriate post-market regulation of<br />

combination products by facilitating the<br />

review process involving more than one<br />

agency center. In this case, the<br />

investigational device exemption (IDE)<br />

application would be sent to the FDA’s<br />

Center for Devices <strong>and</strong> Radiological<br />

Health (CDRH) with significant<br />

consultation by the Office of New <strong>Drug</strong><br />

Quality Assessment (ONDQA)/Division<br />

of Cardiovascular <strong>and</strong> Renal Products.<br />

Many scientific <strong>and</strong> regulatory issues<br />

will arise due to the complex nature of a<br />

coronary DES. Some important concerns<br />

that would need to be evaluated <strong>and</strong><br />

discussed in a submission include acute<br />

<strong>and</strong> chronic stent biocompatibility,<br />

polymeric coating characterization (ie,<br />

thickness, uniformity, integrity, adhesion<br />

to stent), <strong>and</strong> drug release profile. The<br />

predominant percentage composition of a<br />

combination product does not dictate the<br />

Agency center(s) where it is regulated,<br />

rather, it is the primary mode of action or<br />

the “most important therapeutic action of<br />

a combination product” that determines<br />

where a combination product will be<br />

regulated. For example, DES submissions<br />

are assigned to CDRH because the<br />

device’s role in physically maintaining<br />

vessel lumen patency provides the most<br />

important therapeutic action of the<br />

combination product. The drug plays only<br />

a secondary role in reducing restenosis or<br />

re-narrowing of the coronary arteries, a<br />

phenomenon which is caused by the<br />

body’s formation of scar tissue in<br />

response to stent implantation. 12<br />

While the agency currently does not<br />

require the use of Process Analytical<br />

Technologies (PAT) for their submissions,<br />

the use of PAT for CMC documentation<br />

is looked upon favorably. PAT uses tools,<br />

such as near-infrared (NIR) or Raman<br />

spectroscopy, to provide real-time process<br />

data. PAT finds wide applicability in the<br />

pharmaceutical industry <strong>and</strong> can be used<br />

to assess blend <strong>and</strong> content uniformity,<br />

prediction of dissolution time, <strong>and</strong> can<br />

determine the end-point of a coating or<br />

drying operation. Not only can endproduct<br />

testing be reduced, which can<br />

save a company money, but the FDA can<br />

also provide some regulatory flexibility<br />

for any process changes that could occur<br />

in the future, provided that they are<br />

accompanied by scientific justification. 13<br />

“The Agency considers PAT to be a<br />

tool for designing, analyzing, <strong>and</strong><br />

controlling manufacturing through timely<br />

measurements (ie, during processing) of<br />

critical quality <strong>and</strong> performance attributes<br />

of raw <strong>and</strong> in-process materials <strong>and</strong><br />

processes, with the goal of ensuring final<br />

product quality. The goal of PAT is to<br />

enhance underst<strong>and</strong>ing <strong>and</strong> control of the<br />

manufacturing process <strong>and</strong> to facilitate<br />

innovation in development,<br />

manufacturing, <strong>and</strong> quality assurance by<br />

focusing on process underst<strong>and</strong>ing. These<br />

concepts are applicable to all<br />

manufacturing situations,” says Jon E.<br />

Clark, MS, Associate Director for the<br />

Office of Pharmaceutical Science (OPS)<br />

Policy Development at the FDA CDER.<br />

Due to the serious nature of the<br />

diseases they treat, both cardiology <strong>and</strong><br />

oncology drugs qualify under the fasttrack<br />

drug development program<br />

classification. The three designations<br />

given by the FDA are accelerated<br />

approval, fast-track, <strong>and</strong> priority<br />

review. 14,15 In 1992, accelerated approval<br />

for oncology drugs was codified into law<br />

under Subpart H (21 CFR part 314.530)<br />

<strong>and</strong> added to the new drug application<br />

regulations. Accelerated approvals are<br />

granted for the treatment of serious or<br />

life-threatening conditions <strong>and</strong> a benefit<br />

over available therapy exists.<br />

This designation requires using a<br />

surrogate endpoint for efficacy, ie, an<br />

evaluation intended to substitute for a<br />

clinical endpoint such as survival or<br />

morbidity, <strong>and</strong> which is reasonably likely<br />

to predict clinical benefit. Examples of<br />

tumor assessment endpoints are response<br />

rate to drug therapy or time to tumor<br />

progression, measured using anatomic<br />

imaging techniques. Once accelerated<br />

approval has been granted, continued<br />

marketing of the product will be<br />

contingent on the sponsor’s providing<br />

timely <strong>and</strong> conclusive evidence from<br />

validation trials that establishes that the<br />

experimental drug is safe <strong>and</strong> provides<br />

tangible clinical benefit. The product<br />

approval can be withdrawn if<br />

confirmatory studies fail to show clinical<br />

benefits, or if the drug sponsor fails to<br />

conduct the confirmatory studies.<br />

A fast-track status is granted by the<br />

Agency for those drugs also developed to<br />

treat life-threatening diseases <strong>and</strong> that<br />

demonstrate the potential to address an<br />

unmet medical need. In this instance, a<br />

rolling NDA can be approved that allows<br />

for completed sections of the NDA to be<br />

submitted to the FDA on an ongoing<br />

basis. The FDA strongly recommends<br />

several meetings, including a pre-IND<br />

consultation, meetings following Phases<br />

I-II, <strong>and</strong> a pre-NDA meeting to expedite<br />

the approval process. The other<br />

designation, priority review, is for drugs<br />

showing a significant therapeutic benefit<br />

compared to the st<strong>and</strong>ard of care. In this<br />

case, the FDA would review a NDA<br />

within 6 months as opposed to the<br />

st<strong>and</strong>ard review completion timeline of<br />

10 months.<br />

Outsourcing Key<br />

Capabilities<br />

In addition to hearing the term CRO<br />

as a common buzzword these days, the<br />

fact is that both small <strong>and</strong> large<br />

pharmaceutical companies can benefit<br />

from utilizing the added services <strong>and</strong><br />

expertise of a global CRO while focusing<br />

on their core competencies. The CRO<br />

SPECIALTY PHARMA MARCH <strong>2009</strong> Vol 9 No 3<br />

71


SPECIALTY PHARMA MARCH <strong>2009</strong> Vol 9 No 3<br />

72<br />

market size is currently estimated at about $15 billion in revenue per year <strong>and</strong> is<br />

growing at a healthy annual rate of 14.8%. By 2010, the market is forecast to be in<br />

the vicinity of $22.9 billion, according to Frost & Sullivan. A CRO should be chosen<br />

carefully at the outset of a project after considering the budget, timelines, the range<br />

of services provided, <strong>and</strong> the resources offered. A CRO can assist with the review <strong>and</strong><br />

editing of the regulatory submissions <strong>and</strong> can either represent the company as their<br />

sole agent or accompany their client in meetings with the FDA. For smaller<br />

companies with limited resources, limited regulatory expertise, <strong>and</strong> tight timelines,<br />

meetings with the FDA early in the development process can alert the company if<br />

they are going down the wrong pathway.<br />

Meetings with the FDA can be arranged at both the pre-IND stage, where the<br />

company can ask questions to see if they have done enough work to prove a drug’s<br />

safety, <strong>and</strong> also before going through the time <strong>and</strong> expense for Phase III clinical trials<br />

(an after Phase II meeting). These meetings are invaluable in identifying additional<br />

safety, toxicity, or efficacy issues pertinent to cardiology <strong>and</strong> oncology IND/NDA<br />

filings. More frequent interaction with the FDA would be recommended (ie, end of<br />

Phase I meeting) if the drug would be marketed to a small patient population with a<br />

rare disease or condition, ie, orphan drug classification for a disease affecting less<br />

than 200,000 people in the US. Orphan drugs would also be granted accelerated<br />

approval status, <strong>and</strong> NDAs are given a priority review timeline of 6 months.<br />

A CRO can act as a regulatory resource for a company to guide them as to what<br />

the minimum agency requirements are to prove safety <strong>and</strong> efficacy <strong>and</strong> which tests<br />

would be superfluous. Another key advantage of using a CRO is that they have the<br />

capability to offer their clients a strategic viewpoint on risk assessment <strong>and</strong><br />

management, <strong>and</strong> can additionally shoulder some of that risk in interactions with the<br />

FDA. The Agency will take a global view of the scientific data to assess the riskbenefit<br />

ratio, ie, the benefits of the drug substance <strong>and</strong> final drug product must far<br />

outweigh any complications or potential risks to human health. The Agency also<br />

examines what therapeutic advantages the new product has over therapies currently in<br />

the market. Sometimes there is no information available on comparator products as<br />

the regulatory submission is for a first-in-class therapy. In such instances, a CRO can<br />

provide key information as it can draw from a wide knowledge base from past client<br />

experiences with a variety of dosage forms. ◆<br />

References<br />

1. Rosamond W, et al. Heart Disease <strong>and</strong> Stroke Statistics 2008 Update: A Report from the American Heart Association Statistics Committee <strong>and</strong> Stroke Statistics<br />

Subcommittee. Circulation. [Available at: http://circ.ahajournals.org/cgi/content/full/117/4/e25 (DOI: 10.1161/circulationaha.107.187998); Accessed August 6, 2008].<br />

2. Buono D. IMS study finds decline in prescription drug market growth. <strong>Drug</strong> Store News. (April 21, 2008) [Available at:<br />

http://findarticles.com/p/articles/mi_m3374/is_5_30/ai_n25407255; Accessed <strong>July</strong> 30, 2008].<br />

3. Lawrence S. Billion dollar babies-biotech drugs as blockbusters. Nature Biotechnol. 2007;25(4):380-382. [Available at: http://www.nature.com/naturebiotechnology ;<br />

Accessed August 6, 2008].<br />

4. FDA Guidance for Industry M3: Nonclinical Safety Studies for the Conduct of Human Clinical Trials for Pharmaceuticals. Center for <strong>Drug</strong> Evaluation <strong>and</strong> Research<br />

(CDER) & Center for Biologics Evaluation <strong>and</strong> Research (CBER), (<strong>July</strong> 1997).<br />

5. 2008 Report: Medicines in Development for Cancer. [Available at: http://www.phrma.org/files/meds_in_dev/Cancer2008.pdf; Accessed <strong>July</strong> 27, 2008].<br />

6. 2007 Report: Medicines in Development for Heart Disease <strong>and</strong> Stroke. [Available at: http://www.phrma.org/files/Heart2007.pdf; Accessed <strong>July</strong> 27, 2008]<br />

7. FDA Guidance for Industry M4: The CTD- Quality Questions <strong>and</strong> Answers/Location Issues. CDER & CBER, (June 2004).<br />

8. FDA Guidance for Industry M4: The CTD- General Questions <strong>and</strong> Answers. CDER & CBER, (December 2004).<br />

9. FDA Guidance for Industry: Content <strong>and</strong> Format of Investigational New <strong>Drug</strong> Applications (INDs) for Phase 1 Studies of <strong>Drug</strong>s, Including Well-Characterized,<br />

Therapeutic, Biotechnology-derived Products. CDER & CBER, (November 1995).<br />

10. FDA Guidance for Industry: INDs for Phase 2 <strong>and</strong> Phase 3 Studies. Chemistry, Manufacturing, <strong>and</strong> Controls Information. CDER, (May 2003).<br />

11. FDA Guidance for Industry: Applications covered by Section 505(b)(2). CDER (October 1999).<br />

12. FDA Guidance for Industry: Coronary <strong>Drug</strong>-eluting stents- Nonclinical <strong>and</strong> clinical studies. Center for Devices <strong>and</strong> Radiological Health (CDRH) <strong>and</strong> CDER (March<br />

2008).<br />

13. FDA Guidance for Industry: Q8 Pharmaceutical Development. CDER & CBER (May 2006).<br />

14. FDA Guidance for Industry: Fast Track Development Programs- Designation, Development, <strong>and</strong> Application Review. CDER & CBER (January 2006).<br />

15. Dagher R, Johnson J, Williams G, Keegan P, Pazdur R. Accelerated Approval of <strong>Oncology</strong> Products: A Decade of Experience. J Natl Cancer Inst. 2004;96(20):1500-<br />

1509.<br />

Stuart Cantor,<br />

PhD<br />

Senior Scientist<br />

<strong>ICON</strong> Development<br />

Solutions<br />

Dr. Stuart Cantor graduated from the University of<br />

Maryl<strong>and</strong> Pharmacy School <strong>and</strong> is a Senior Scientist<br />

for <strong>ICON</strong>. Dr. Cantor currently assists global clients<br />

with their CMC sections of regulatory documents for<br />

solid dosage forms <strong>and</strong> biologics, <strong>and</strong> also h<strong>and</strong>les<br />

clinical supply chain management issues. He has 7<br />

pharmaceutical publications online/in-process, <strong>and</strong><br />

has published a book chapter on wet granulation.<br />

He interned at Wyeth <strong>and</strong> Bristol Myers Squibb<br />

(BMS) <strong>and</strong> studied different granulation processes<br />

<strong>and</strong> their mechanical properties. His research<br />

covered preformulation, formulation, analytical<br />

method validation, blend segregation, <strong>and</strong> chemical<br />

imaging. His expertise is in extrusionspheronization,<br />

wet granulation, <strong>and</strong> NIR<br />

spectroscopy. Dr. Cantor previously developed the<br />

sugarless fiber chews for diabetics launched by<br />

BMS under the Choice ® DM br<strong>and</strong>.<br />

Kadriye Ciftci,<br />

PhD<br />

Senior Director <strong>Drug</strong><br />

Delivery<br />

<strong>ICON</strong> Development<br />

Solutions<br />

Dr. Kadriye Çiftci is Senior Director of <strong>Drug</strong><br />

Delivery/Formulations at <strong>ICON</strong> Development<br />

Solutions. She has more than 15 years of<br />

experience in academia <strong>and</strong> pharmaceutical R&D.<br />

Dr. Ciftci completed her training at the University of<br />

Illinois at Chicago <strong>and</strong> the University of Michigan<br />

Medical School. She previously worked as a Tenuretrack<br />

Assistant Professor at Temple University <strong>and</strong> a<br />

Research Fellow at the Schering Plough Research<br />

Institute. Her special interests include cancer<br />

research, gene therapy, <strong>and</strong> the development of<br />

novel drug delivery systems, particularly biotech<br />

products <strong>and</strong> vaccines.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!