31.05.2022 Views

v39i2

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

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

Volume 39 Issue 2

June 2022

E-ISSN: 1308-5263

Research Articles

Comprehensive Mutation Profile in Acute Myeloid Leukemia Patients with RUNX1-RUNX1T1 or CBFB-MYH11

Fusions

Wei Qin, Xiayu Chen, Hong Jie Shen, Zheng Wang, Xiaohui Cai, Naike Jiang, Haiying Hua; Changzhou, Wuxi,

Soochow, Suzhou, China

Invasive Fungal Infections in Children with Leukemia: Clinical Features and Prognosis

Melike Sezgin Evim, Özlem Tüfekçi, Birol Baytan, Hale Ören, Solmaz Çelebi, Beyza Ener, Kevser Üstün Elmas,

Şebnem Yılmaz, Melek Erdem, Mustafa Kemal Hacımustafaoğlu, Adalet Meral Güneş; Bursa, İzmir, Turkey

Eltrombopag for Treatment of Thrombocytopenia Following Hematopoietic Stem Cell Transplantation

Zeynep Tuğba Güven, Serhat Çelik, Bülent Eser, Mustafa Çetin, Ali Ünal, Leylagül Kaynar; Kayseri, Antalya,

Turkey

Assessment of Bone Marrow Biopsy and Cytogenetic Findings in Patients with Multiple Myeloma

Ahmet Şeyhanlı, Boran Yavuz, Zehra Akşit, Zeynep Yüce, Sermin Özkal, Oğuz Altungöz, Fatih Demirkan,

İnci Alacacıoğlu, Güner Hayri Özsan; Sivas, İzmir, Turkey

Combination of Haploidentical Hematopoietic Stem Cell Transplantation with Umbilical Cord-Derived

Mesenchymal Stem Cells in Patients with Severe Aplastic Anemia: A Retrospective Controlled Study

Xian-Fu Sheng, Hui Li, Li-Li Hong, Hai-Feng Zhuang; Zhejiang, China

Castleman Disease: A Multicenter Case Series from Turkey

Eren Gündüz, Hakkı Onur Kırkızlar, Elif Gülsüm Ümit, Sedanur Karaman Gülsaran, Vildan Özkocaman,

Fahir Özkalemkaş, Ömer Candar, Tugrul Elverdi, Selin Küçükyurt, Semra Paydaş, Özcan Çeneli, Sema Karakuş,

Senem Maral, Ömer Ekinci, Yıldız İpek, Cem Kis, Zeynep Tuğba Güven, Aydan Akdeniz, Tiraje Celkan, Ayşe Hilal

Eroğlu Küçükdiler, Gülsüm Akgün Çağlıyan, Ceyda Özçelik Şengöz, Ayşe Karataş, Tuba Bulduk, Alper Özcan,

Fatma Burcu Belen Apak, Aylin Canbolat, İbrahim Kartal, Hale Ören, Ersin Töret, Gül Nihal Özdemir, Şule Mine

Bakanay Öztürk; Eskişehir, Edirne, Bursa, İstanbul, Adana, Konya, Ankara, Elazığ, Kayseri, Mersin, Aydın, Denizli,

Trabzon, Samsun, İzmir, Turkey

Cover Picture:

Gül Nihal Özdemir, Eren Gündüz

Gaucher Disease for Hematologists

2


Editor-in-Chief

Reyhan Küçükkaya

İstanbul, Turkey

rkucukkaya@hotmail.com

Associate Editors

Cengiz Beyan

Retired Professor

cengizbeyan@hotmail.com

A. Emre Eşkazan

İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine,

Department of Internal Medicine, Division of Hematology, İstanbul, Turkey

emre.eskazan@istanbul.edu.tr

Hale Ören

Dokuz Eylül University Faculty of Medicine, Department of Pediatric

Hematology, İzmir, Turkey

hale.oren@deu.edu.tr

Ali İrfan Emre Tekgündüz

Memorial Bahçelievler Hospital, Adult Hematology and BMT Clinic,

İstanbul, Turkey

emretekgunduz@yahoo.com

Şule Ünal

Hacettepe University Faculty of Medicine, Division of Pediatric Hematology,

Ankara, Turkey

suleunal2003@hotmail.com

Ayşegül Ünüvar

İstanbul University, İstanbul School of Medicine, Division of Pediatric

Hematology-Oncology, İstanbul, Turkey

aysegulu@hotmail.com

Assistant Editors

Olga Meltem Akay

Koç University Faculty of Medicine, Department of Hematology,

İstanbul, Turkey

İnci Alacacıoğlu

Dokuz Eylül University, Institute of Oncology, Clinical Oncology Department,

Division of Hematology, İzmir, Turkey

Claudio Cerchione

Scientific Institute of Romagna for the Study and Treatment of Tumors,

Unit of Hematology, Meldola, Italy

Nil Güler

Pamukkale University Faculty of Medicine, Department of Hematology,

Denizli, Turkey

Veysel Sabri Hançer

İstinye University Faculty of Medicine, Department of Medical Biology,

İstanbul, Turkey

Elif Ünal İnce

Ankara University Faculty of Medicine, Department of Pediatric Hematology,

Ankara, Turkey

Zühre Kaya

Gazi University Faculty of Medicine, Department of Hematology, Ankara,

Turkey

Ebru Koca

Başkent University Ankara Hospital, Clinic of Hematology, Ankara, Turkey

Müge Sayitoğlu

İstanbul University, Aziz Sancar Institute of Experimental Medicine,

Department of Genetics, İstanbul, Turkey

Mario Tiribelli

University of Udine, Department of Medical Area, Division of Hematology

and Stem Cell Transplantation, Udine, Italy

International Advisory Board

Nejat Akar (Turkey)

TOBB University of Economics and Technology, Clinic of Child Health and

Diseases (Pediatrics), Ankara, Turkey

Görgün Akpek (USA)

Dignity Health Center San Francisco, California, USA

Serhan Alkan (USA)

Loyola University Medical Center, Department of Pathology, IL, USA

Meral Beksaç (Turkey)

Ankara University Faculty of Medicine, Department of Hematology, Ankara,

Turkey

Koen van Besien (USA)

Weill Cornell Medical College, New York, USA

M. Sıraç Dilber (Sweden)

Karolinska University Hospital Huddinge, Stockholm, Sweden

Ahmet Doğan (USA)

Memorial Sloan Kettering Cancer Center, New York, USA

Peter Dreger (Germany)

Heidelberg University Hospital, Department of Internal Medicine V,

Heidelberg, Germany

Thierry Facon (France)

Lille University, Department of Hematology, Lille, France

Jawed Fareed (USA)

Loyola University Medical Center, Maywood, IL, USA

Burhan Ferhanoğlu (Turkey)

Koç University Faculty of Medicine, Department of Hematology, İstanbul,

Turkey

Gösta Gahrton (Sweden)

Karolinska Institute at Huddinge and Solna, Department of Medicine,

Stockholm, Sweden

Margarita Guenova (Bulgaria)

National Center of Hematology and Transfusiology, Department of

Morphology, Sofia, Bulgaria

A-I

Publishing

Services

GALENOS PUBLISHER

Molla Gürani Mah. Kaçamak Sk. No: 21/1, Fındıkzade, İstanbul, Turkey

Phone: +90 212 621 99 25 • Fax: +90 212 621 99 27 • www. galenos.com.tr


İbrahim Haznedaroğlu (Turkey)

Hacettepe University Faculty of Medicine, Department of Hematology,

Ankara, Turkey

Dieter Hoelzer (Germany)

University Hospital, Department of Internal Medicine II, Hematology and

Oncology, Frankfurt, Germany

Marilyn Manco-Johnson (USA)

Hemophilia & Thrombosis Center, Section of Hematology, Oncology, and

Bone Marrow Transplantation, Department of Pediatrics, University of

Colorado Denver and The Children’s Hospital, Aurora, CO

Andreas Josting (Germany)

German Hodgkin Study Group (GHSG), Cologne, Germany; Protestant

Hospital Lippstadt, Lippstadt, Germany

Emin Kansu (Turkey)

Retired Professor, Hacettepe University Research Center for Stem Cells

(PEDİ-STEM), Senior Consultant

Winfried Kern (Germany)

University of Freiburg, Faculty of Medicine, University of Freiburg,

Department of Medicine II, Division of Infectious Diseases, Medical

Center, Freiburg, Germany

Nigel Key (USA)

University of Minnesota Medical School, Minneapolis, MN, USA

Selami Koçak Toprak (Turkey)

Ankara University School of Medicine, Department of Hematology,

Ankara, Turkey

Korgün Koral (USA)

University of Texas Southwestern Medical Center at Dallas and

Children’s Medical Center Department of Radiology, Texas, USA

Abdullah Kutlar (USA)

Medical College of Georgia, Department of Medicine, Augusta, GA

Luca Malcovati (Italy)

University of Pavia, Fondazione IRCCS Policlinico San Matteo,

Department of Hematology, Pavia, Italy

Robert Marcus (United Kingdom)

King’s College Hospital, London, United Kingdom

Jean Pierre Marie (France)

Laboratoire de Culture et Cinétique Cellulaire, Hôpital Hôtel-Dieu, Paris,

France

Gerassimos A. Pangalis (Greece)

National and Kapodistrian University of Athens, Laikon General Hospital,

Department of Haematology, Athens, Greece

Semra Paydaş (Turkey)

Çukurova University Faculty of Medicine, Department of Oncology,

Adana, Turkey

Santiago Pavlovsky (Argentina)

FUNDALEU (Foundation to Fight Leukemia) and Centro de Investigacion

Clinica A. Ocampo, Buenos Aires, Argentina

Antonio Giulio Piga (Italy)

University of Turin, Department of Clinical and Biological Sciences,

Torino, Italy

Ananda Prasad (USA)

Wayne State University, School of Medicine, Department of Internal

Medicine, Division of Hematology/Oncology, Detroit, Michigan, USA

Jacob M. Rowe (Israel)

Hebrew University of Jerusalem, Jerusalem, Israel

Jens-Ulrich Rüffer (Germany)

German Fatigue Society, Cologne, Germany

Norbert Schmitz (Germany)

Asklepios Hospital Barmbek, Department of Hematology, Oncology and

Stem Cell Transplantation, Hamburg, Germany

Orhan Sezer (Germany)

Charité Comprehensive Cancer Center, Berlin, Germany

Anna Sureda (Spain)

ICO - Hospital Duran i Reynals, Barcelona, Spain

Ayalew Tefferi (USA)

Mayo Clinic, Divisions of Hematology, Rochester, Minnesota, USA

Nüket Tüzüner (Turkey)

Retired Professor

Catherine Verfaillie (Belgium)

Katholieke Universiteit Leuven, Leuven, Belgium

Srdan Verstovsek (USA)

UT MD Anderson Cancer Center, Department of Leukemia, Houston, TX,

USA

Claudio Viscoli (Italy)

IRCCS San Martino-IST, University of Genoa, Clinic of Infectious

Diseases, Genoa, Italy

Past Editors

Erich Frank

Orhan Ulutin

Hamdi Akan

Aytemiz Gürgey

Senior Advisory Board

Yücel Tangün

Osman İlhan

Muhit Özcan

Teoman Soysal

Ahmet Muzaffer Demir

Güner Hayri Özsan

Language Editor

Leslie Demir

Statistic Editor

Hülya Ellidokuz

Editorial Office

İpek Durusu

Bengü Timoçin Efe

A-II


Contact Information

Editorial Correspondence should be addressed to Dr. Reyhan Küçükkaya

E-mail : rkucukkaya@hotmail.com

All Inquiries Should be Addressed to

TURKISH JOURNAL OF HEMATOLOGY

Address : Turan Güneş Bulv. İlkbahar Mah. Fahreddin Paşa Sokağı (eski 613. Sok.) No: 8 06550 Çankaya, Ankara / Turkey

Phone : +90 312 490 98 97

Fax : +90 312 490 98 68

E-mail : tjh@tjh.com.tr

E-ISSN: 1308-5263

Publishing Manager

Reyhan Küçükkaya

Management Address

Türk Hematoloji Derneği

Turan Güneş Bulv. İlkbahar Mah. Fahreddin Paşa Sokağı (eski 613. Sok.)

No: 8 06550 Çankaya, Ankara / Turkey

Online Manuscript Submission

http://mc.manuscriptcentral.com/tjh

Web Page

www.tjh.com.tr

Owner on Behalf of the Turkish Society

of Hematology

Muhlis Cem Ar

Publishing House

Molla Gürani Mah. Kaçamak Sk. No: 21,

34093 Fındıkzade, İstanbul / Turkey

Tel: +90 212 621 99 25

Fax: +90 212 621 99 27

E-mail: info@galenos.com.tr

Publisher Certificate Number: 14521

Publication Date

01.06.2022

Cover Picture

Gül Nihal Özdemir, Eren Gündüz

Gaucher Disease for Hematologists

Bone marrow aspirate showing a number of large

macrophages laden with cerebrosides (arrows: Gaucher cells) in a

patient with Gaucher disease.

International scientific journal published quarterly.

The Turkish Journal of Hematology is published by the commercial enterprise

of the Turkish Society of Hematology with Decision Number 6 issued by the

Society on 7 October 2008.

A-III


AIMS AND SCOPE

The Turkish Journal of Hematology is published quarterly (March, June,

September, and December) by the Turkish Society of Hematology. It is an

independent, non-profit peer-reviewed international English-language

periodical encompassing subjects relevant to hematology.

The Editorial Board of The Turkish Journal of Hematology adheres to the

principles of the World Association of Medical Editors (WAME), International

Council of Medical Journal Editors (ICMJE), Committee on Publication

Ethics (COPE), Consolidated Standards of Reporting Trials (CONSORT) and

Strengthening the Reporting of Observational Studies in Epidemiology

(STROBE).

The aim of The Turkish Journal of Hematology is to publish original

hematological research of the highest scientific quality and clinical relevance.

Additionally, educational material, reviews on basic developments, editorial

short notes, images in hematology, and letters from hematology specialists

and clinicians covering their experience and comments on hematology and

related medical fields as well as social subjects are published. As of December

2015, The Turkish Journal of Hematology does not accept case reports.

General practitioners interested in hematology and internal medicine

specialists are among our target audience, and The Turkish Journal of

Hematology aims to publish according to their needs. The Turkish Journal of

Hematology is indexed, as follows:

- PubMed Medline

- PubMed Central

- Science Citation Index Expanded

- EMBASE

- Scopus

- CINAHL

- Gale/Cengage Learning

- EBSCO

- ProQuest

- Index Copernicus

- Tübitak/Ulakbim Turkish Medical Database

- Turk Medline

- Hinari

- GOALI

- ARDI

- OARE

Impact Factor: 1.831

Digital Archiving Policy

The journal’s Abstracting/Indexing services store essential information about

articles. In addition, some of our journal’s Abstracting/Indexing services

archive metadata about the article and electronic versions of the articles.

In this way, copies of articles are presented to the scientific community

through these systems as an alternative to journals.

Open Access Policy

This journal provides immediate open access to its content on the principle

that making research freely available to the public supports a greater global

exchange of knowledge. Open Access Policy is based on rules of Budapest

Open Access Initiative (BOAI) http://www.budapestopenaccessinitiative.org/.

By “open access” to [peer-reviewed research literature], we mean its free

availability on the public internet, permitting any users to read, download,

copy, distribute, print, search, or link to the full texts of these articles, crawl

them for indexing, pass them as data to software, or use them for any

other lawful purpose, without financial, legal, or technical barriers other

than those inseparable from gaining access to the internet itself. The only

constraint on reproduction and distribution, and the only role for copyright

in this domain, should be to give authors control over the integrity of their

work and the right to be properly acknowledged and cited.

This work is licensed under a Creative Commons Attribution-NonCommercial-

NoDerivatives 4.0 (CC BY-NC-ND) International License.

CC BY-NC-ND: This license allows reusers to copy and distribute the material

in any medium or format in unadapted form only, for noncommercial

purposes only, and only so long as attribution is given to the creator.

CC BY-NC-ND includes the following elements:

BY – Credit must be given to the creator

NC – Only noncommercial uses of the work are permitted

ND – No derivatives or adaptations of the work are permitted

Permission Requests

Permission is required for the use of any work published under a Creative

Commons Attribution-NonCommercial-NoDerivatives 4.0 (CC BY-NC-ND)

International License with commercial purposes (selling, etc.) to protect

the copyright owner and author rights. Republication and reproduction

of images or tables in any published material should be done with proper

citation of source providing authors’ names; article title; journal title; year

(volume) and page of publication; copyright year of the article.

Subscription Information

The Turkish Journal of Hematology is published electronically only as of

2019. Therefore, subscriptions are not necessary. All published volumes are

available in full text free-of-charge online at www.tjh.com.tr.

Address: Turan Güneş Bulv. İlkbahar Mah. Fahreddin Paşa Sokağı (eski 613.

Sok.) No: 8 06550 Çankaya, Ankara / Turkey

Telephone: +90 312 490 98 97

Fax: +90 312 490 98 68

Online Manuscript Submission: http://mc.manuscriptcentral.com/tjh

Web page: www.tjh.com.tr

E-mail: tjh@tjh.com.tr

Permissions

Requests for permission to reproduce published material should be sent to

the editorial office.

Editor: Professor Dr. Reyhan Küçükkaya

Adress: Turan Güneş Bulv. İlkbahar Mah. Fahreddin Paşa Sokağı (eski 613.

Sok.) No: 8 06550 Çankaya, Ankara / Turkey

Telephone: +90 312 490 98 97

Fax: +90 312 490 98 68

Online Manuscript Submission: http://mc.manuscriptcentral.com/tjh

Web page: www.tjh.com.tr

E-mail: tjh@tjh.com.tr

A-IV


Publisher

Galenos Yayınevi

Molla Gürani Mah. Kaçamak Sk. No:21 34093 Fındıkzade-İstanbul, Turkey

Telephone : +90 212 621 99 25

Fax : +90 212 621 99 27

info@galenos.com.tr

Instructions for Authors

Instructions for authors are published in the journal and at www.tjh.com.tr

Material Disclaimer

Authors are responsible for the manuscripts they publish in The Turkish

Journal of Hematology. The editor, editorial board, and publisher do not

accept any responsibility for published manuscripts.

If you use a table or figure (or some data in a table or figure) from another

source, cite the source directly in the figure or table legend.

Editorial Policy

Following receipt of each manuscript, a checklist is completed by the

Editorial Assistant. The Editorial Assistant checks that each manuscript

contains all required components and adheres to the author guidelines, after

which time it will be forwarded to the Editor in Chief. Following the Editor

in Chief’s evaluation, each manuscript is forwarded to the Associate Editor,

who in turn assigns reviewers. Generally, all manuscripts will be reviewed

by at least three reviewers selected by the Associate Editor, based on their

relevant expertise. The Associate Editor could be assigned as a reviewer

along with the other reviewers. After the reviewing process, all manuscripts

are evaluated in the Editorial Board Meeting.

The Turkish Journal of Hematology’s editors and Editorial Board members

are active researchers. It is possible that they may desire to submit their

manuscripts to the Turkish Journal of Hematology. This may create a conflict

of interest. These manuscripts will not be evaluated by the submitting

editor(s). The review process will be managed and decisions made by the

Editor in Chief, who will act independently. In some situations, this process

will be overseen by an outside independent expert in reviewing submissions

from editors.

A-V


TURKISH JOURNAL OF HEMATOLOGY

INSTRUCTIONS FOR AUTHORS

The Turkish Journal of Hematology accepts invited review articles,

research articles, brief reports, letters to the editor, and hematological

images that are relevant to the scope of hematology, on the condition

that they have not been previously published elsewhere. Basic science

manuscripts, such as randomized, cohort, cross-sectional, and casecontrol

studies, are given preference. All manuscripts are subject

to editorial revision to ensure they conform to the style adopted by

the journal. There is a double-blind reviewing system. Review articles

are solicited by the Editor-in-Chief. Authors wishing to submit an

unsolicited review article should contact the Editor-in-Chief prior to

submission in order to screen the proposed topic for relevance and

priority.

The Turkish Journal of Hematology does not charge any article

submission or processing charges.

Manuscripts should be prepared according to ICMJE guidelines (http://

www.icmje.org/). Original manuscripts require a structured abstract.

Label each section of the structured abstract with the appropriate

subheading (Objective, Materials and Methods, Results, and Conclusion).

Letters to the editor do not require an abstract. Research or project

support should be acknowledged as a footnote on the title page.

Technical and other assistance should be provided on the title page.

Submissions and publication are free of charge.

Original Manuscripts

Title Page

Title: The title should provide important information regarding the

manuscript’s content. The title must specify that the study is a cohort

study, cross-sectional study, case-control study, or randomized study (i.e.

Cao GY, Li KX, Jin PF, Yue XY, Yang C, Hu X. Comparative bioavailability

of ferrous succinate tablet formulations without correction for baseline

circadian changes in iron concentration in healthy Chinese male

subjects: A single-dose, randomized, 2-period crossover study. Clin Ther

2011;33:2054-2059).

The title page should include the authors’ names, degrees, and

institutional/professional affiliations and a short title, abbreviations,

keywords, financial disclosure statement, and conflict of interest

statement. If a manuscript includes authors from more than one

institution, each author’s name should be followed by a superscript

number that corresponds to their institution, which is listed separately.

Please provide contact information for the corresponding author,

including name, e-mail address, and telephone and fax numbers.

Important Notice: The title page should be submitted separately.

Running Head: The running head should not be more than 40

characters, including spaces, and should be located at the bottom of

the title page.

Word Count: A word count for the manuscript, excluding abstract,

acknowledgments, figure and table legends, and references, should be

provided and should not exceed 2500 words. The word count for the

abstract should not exceed 300 words.

Conflict of Interest Statement: To prevent potential conflicts of

interest from being overlooked, this statement must be included in each

manuscript. In case there are conflicts of interest, every author should

complete the ICMJE general declaration form, which can be obtained at

http://www.icmje.org/downloads/coi_disclosure.zip

Abstract and Keywords: The second page should include an abstract

that does not exceed 300 words. For manuscripts sent by authors in

Turkey, a title and abstract in Turkish are also required. As most readers

read the abstract first, it is critically important. Moreover, as various

electronic databases integrate only abstracts into their index, important

findings should be presented in the abstract.

Objective: The abstract should state the objective (the purpose of the

study and hypothesis) and summarize the rationale for the study.

Materials and Methods: Important methods should be written

respectively.

Results: Important findings and results should be provided here.

Conclusion: The study’s new and important findings should be

highlighted and interpreted.

Other types of manuscripts, such as reviews, brief reports, and

editorials, will be published according to uniform requirements.

Provide 3-10 keywords below the abstract to assist indexers. Use

terms from the Index Medicus Medical Subject Headings List

(for randomized studies a CONSORT abstract should be provided: http://

www.consort-statement.org).

Introduction: The introduction should include an overview of the

relevant literature presented in summary form (one page), and whatever

remains interesting, unique, problematic, relevant, or unknown about

the topic must be specified. The introduction should conclude with the

rationale for the study, its design, and its objective(s).

Materials and Methods: Clearly describe the selection of observational

or experimental participants, such as patients, laboratory animals, and

controls, including inclusion and exclusion criteria and a description of

the source population. Identify the methods and procedures in sufficient

detail to allow other researchers to reproduce your results. Provide

references to established methods (including statistical methods),

provide references to brief modified methods, and provide the rationale

for using them and an evaluation of their limitations. Identify all drugs

and chemicals used, including generic names, doses, and routes of

administration. The section should include only information that was

available at the time the plan or protocol for the study was devised

A-VI


(https://www.strobe-statement.org/fileadmin/Strobe/uploads/checklists/

STROBE_checklist_v4_combined.pdf).

Statistics: Describe the statistical methods used in enough detail to

enable a knowledgeable reader with access to the original data to verify

the reported results. Statistically important data should be given in the

text, tables, and figures. Provide details about randomization, describe

treatment complications, provide the number of observations, and specify

all computer programs used.

Results: Present your results in logical sequence in the text, tables, and

figures. Do not present all the data provided in the tables and/or figures

in the text; emphasize and/or summarize only important findings, results,

and observations in the text. For clinical studies provide the number of

samples, cases, and controls included in the study. Discrepancies between

the planned number and obtained number of participants should be

explained. Comparisons and statistically important values (i.e. p-value

and confidence interval) should be provided.

Discussion: This section should include a discussion of the data. New and

important findings/results and the conclusions they lead to should be

emphasized. Link the conclusions with the goals of the study, but avoid

unqualified statements and conclusions not completely supported by the

data. Do not repeat the findings/results in detail; important findings/results

should be compared with those of similar studies in the literature, along with

a summarization. In other words, similarities or differences in the obtained

findings/results with those previously reported should be discussed.

Study Limitations: Limitations of the study should be detailed. In

addition, an evaluation of the implications of the obtained findings/

results for future research should be outlined.

Conclusion: The conclusion of the study should be highlighted.

References

Cite references in the text, tables, and figures with numbers in square

brackets. Number references consecutively according to the order in

which they first appear in the text. Journal titles should be abbreviated

according to the style used in Index Medicus (consult List of Journals

Indexed in Index Medicus). Include among the references any paper

accepted, but not yet published, designating the journal followed by “in

press”.

Examples of References:

1. List all authors

Deeg HJ, O’Donnel M, Tolar J. Optimization of conditioning for marrow

transplantation from unrelated donors for patients with aplastic anemia

after failure of immunosuppressive therapy. Blood 2006;108:1485-1491.

2. Organization as author

Royal Marsden Hospital Bone Marrow Transplantation Team. Failure of

syngeneic bone marrow graft without preconditioning in post-hepatitis

marrow aplasia. Lancet 1977;2:742-744.

3. Book

Wintrobe MM. Clinical Hematology, 5th ed. Philadelphia, Lea & Febiger, 1961.

4. Book Chapter

Perutz MF. Molecular anatomy and physiology of hemoglobin. In:

Steinberg MH, Forget BG, Higs DR, Nagel RI, (eds). Disorders of Hemoglobin:

Genetics, Pathophysiology, Clinical Management. New York, Cambridge

University Press, 2000.

5. Abstract

Drachman JG, Griffin JH, Kaushansky K. The c-Mpl ligand (thrombopoietin)

stimulates tyrosine phosphorylation. Blood 1994;84:390a (abstract).

6. Letter to the Editor

Rao PN, Hayworth HR, Carroll AJ, Bowden DW, Pettenati MJ. Further

definition of 20q deletion in myeloid leukemia using fluorescence in situ

hybridization. Blood 1994;84:2821-2823.

7. Supplement

Alter BP. Fanconi’s anemia, transplantation, and cancer. Pediatr Transplant

2005;9(Suppl 7):81-86.

Brief Reports

Abstract length: Not to exceed 150 words.

Article length: Not to exceed 1200 words.

Introduction: State the purpose and summarize the rationale for the study.

Materials and Methods: Clearly describe the selection of the observational

or experimental participants. Identify the methods and procedures in

sufficient detail. Provide references to established methods (including

statistical methods), provide references to brief modified methods, and

provide the rationale for their use and an evaluation of their limitations.

Identify all drugs and chemicals used, including generic names, doses, and

routes of administration.

Statistics: Describe the statistical methods used in enough detail to

enable a knowledgeable reader with access to the original data to verify

the reported findings/results. Provide details about randomization,

describe treatment complications, provide the number of observations,

and specify all computer programs used.

Results: Present the findings/results in a logical sequence in the text, tables,

and figures. Do not repeat all the findings/results in the tables and figures in

the text; emphasize and/or summarize only those that are most important.

Discussion: Highlight the new and important findings/results of the

study and the conclusions they lead to. Link the conclusions with the

goals of the study, but avoid unqualified statements and conclusions not

completely supported by your data.

Invited Review Articles

Abstract length: Not to exceed 300 words.

Article length: Not to exceed 4000 words.

Review articles should not include more than 100 references. Reviews should

include a conclusion, in which a new hypothesis or study about the subject

A-VII


may be posited. Do not publish methods for literature search or level of

evidence. Authors who will prepare review articles should already have

published research articles on the relevant subject. The study’s new and

important findings should be highlighted and interpreted in the Conclusion

section. There should be a maximum of two authors for review articles.

Perspectives in Hematology

“Perspectives” are articles discussing significant topics relevant to

hematology. They are more personal than a Review Article. Authors

wishing to submit a Perspective in Hematology article should contact

the Editor in Chief prior to submission in order to screen the proposed

topic for relevance and priority. Articles submitted for “Perspectives

in Hematology” must advance the hot subjects of experimental and/

or clinical hematology beyond the articles previously published or in

press in TJH. Perspective papers should meet the restrictive criteria of

TJH regarding unique scientific and/or educational value, which will

impact and enhance clinical hematology practice or the diagnostic

understanding of blood diseases. Priority will be assigned to such

manuscripts based upon the prominence, significance, and timeliness of

the content. The submitting author must already be an expert with a

recognized significant published scientific experience in the specific field

related to the “Perspectives” article.

Abstract length: Not to exceed 150 words.

Article length: Not to exceed 1000 words.

References: Should not include more than 50 references

Images in Hematology

Article length: Not to exceed 200 words.

Authors can submit for consideration illustrations or photos that are

interesting, instructive, and visually attractive, along with a few lines

of explanatory text and references. Images in Hematology can include

no more than 200 words of text, 5 references, and 3 figures or tables.

No abstract, discussion, or conclusion is required, but please include a

brief title.

Letters to the Editor

Article length: Not to exceed 500 words.

Letters can include no more than 500 words of text, 5-10 references,

and 1 figure or table. No abstract is required, but please include a brief

title. The total number is usually limited to a maximum of five authors

for a letter to the editor.

Tables

Supply each table in a separate file. Number tables according to the order

in which they appear in the text, and supply a brief caption for each.

Give each column a short or abbreviated heading. Write explanatory

statistical measures of variation, such as standard deviation or standard

error of mean. Be sure that each table is cited in the text.

Figures

Figures should be professionally drawn and/or photographed. Authors

should number figures according to the order in which they appear in

the text. Figures include graphs, charts, photographs, and illustrations.

Each figure should be accompanied by a legend that does not exceed

50 words. Use abbreviations only if they have been introduced in the

text. Authors are also required to provide the level of magnification for

histological slides. Explain the internal scale and identify the staining

method used. Figures should be submitted as separate files, not in

the text file. High-resolution image files are not preferred for initial

submission as the file sizes may be too large. The total file size of the

PDF for peer review should not exceed 5 MB.

Authorship

Each author should have participated sufficiently in the work to assume

public responsibility for the content. Any portion of a manuscript that is

critical to its main conclusions must be the responsibility of at least one

author.

Contributor’s Statement

All submissions should contain a contributor’s statement page. Each

statement should contain substantial contributions to idea and design,

acquisition of data, and analysis and interpretation of findings. All persons

designated as an author should qualify for authorship, and all those that

qualify should be listed. Each author should have participated sufficiently

in the work to take responsibility for appropriate portions of the text.

Acknowledgments

Acknowledge support received from individuals, organizations, grants,

corporations, and any other source. For work involving a biomedical

product or potential product partially or wholly supported by corporate

funding, a note stating, “This study was financially supported (in part)

with funds provided by (company name) to (authors’ initials)”, must

be included. Grant support, if received, needs to be stated and the

specific granting institutions’ names and grant numbers provided when

applicable.

Authors are expected to disclose on the title page any commercial or

other associations that might pose a conflict of interest in connection

with the submitted manuscript. All funding sources that supported the

work and the institutional and/or corporate affiliations of the authors

should be acknowledged on the title page.

Ethics

When reporting experiments conducted with humans indicate that

the procedures were in accordance with ethical standards set forth

by the committee that oversees human subject research. Approval of

research protocols by the relevant ethics committee, in accordance with

international agreements (Helsinki Declaration of 1975, revised 2013

available at https://www.wma.net/policies-post/wma-declaration-ofhelsinki-ethical-principles-for-medical-research-involving-humansubjects/),

is required for all experimental, clinical, and drug studies.

Patient names, initials, and hospital identification numbers should

not be used. Manuscripts reporting the results of experimental

A-VIII


investigations conducted with humans must state that the study protocol

received institutional review board approval and that the participants

provided informed consent.

Non-compliance with scientific accuracy is not in accord with scientific

ethics. Plagiarism: To re-publish, in whole or in part, the contents of

another author’s publication as one’s own without providing a reference.

Fabrication: To publish data and findings/results that do not exist.

Duplication: Use of data from another publication, which includes republishing

a manuscript in different languages. Salami slicing: To create

more than one publication by dividing the results of a study unnecessarily.

We disapprove of such unethical practices as plagiarism, fabrication,

duplication, and salami slicing, as well as efforts to influence the

review process with such practices as gifting authorship, inappropriate

acknowledgments, and references. Additionally, authors must respect

participants‘ right to privacy.

On the other hand, short abstracts published in congress books that do

not exceed 400 words and present data of preliminary research, and those

that are presented in an electronic environment, are not considered as

previously published work. Authors in such a situation must declare this

status on the first page of the manuscript and in the cover letter.

(The COPE flowchart is available at http://publicationethics.org.)

We use iThenticate to screen all submissions for plagiarism before

publication.

Turkish Journal of Hematology uses plagiarism screening service to verify

the originality of content submitted before publication. This journal does

not accept articles that indicate a similarity rate of more than 30%,

according to iThenticate reports.

Conditions of Publication

All authors are required to affirm the following statements before their

manuscript is considered: 1. The manuscript is being submitted only

to The Turkish Journal of Hematology; 2. The manuscript will not be

submitted elsewhere while under consideration by The Turkish Journal

of Hematology; 3. The manuscript has not been published elsewhere,

and should it be published in The Turkish Journal of Hematology it will

not be published elsewhere without the permission of the editors (these

restrictions do not apply to abstracts or to press reports for presentations

at scientific meetings); 4. All authors are responsible for the manuscript’s

content; 5. All authors participated in the study concept and design,

analysis and interpretation of the data, and drafting or revising of the

manuscript and have approved the manuscript as submitted. In addition,

all authors are required to disclose any professional affiliation, financial

agreement, or other involvement with any company whose product

figures prominently in the submitted manuscript.

Authors of accepted manuscripts will receive electronic page proofs and

are responsible for proofreading and checking the entire article within

two days. Failure to return the proof in two days will delay publication.

If the authors cannot be reached by email or telephone within two

weeks, the manuscript will be rejected and will not be published in the

journal.

Copyright

At the time of submission all authors will receive instructions for submitting

an online copyright form. No manuscript will be considered for review until

all authors have completed their copyright form. Please note, it is our practice

not to accept copyright forms via fax, e-mail, or postal service unless there

is a problem with the online author accounts that cannot be resolved. Every

effort should be made to use the online copyright system. Corresponding

authors can log in to the submission system at any time to check the status

of any co-author’s copyright form. All accepted manuscripts become the

permanent property of The Turkish Journal of Hematology and may not be

published elsewhere, in whole or in part, without written permission.

Note: We cannot accept any copyright form that has been altered,

revised, amended, or otherwise changed. Our original copyright form

must be used as is.

Units of Measurement

Measurements should be reported using the metric system, according

to the International System of Units (SI). Consult the SI Unit Conversion

Guide, New England Journal of Medicine Books, 1992.

An extensive list of conversion factors can be found at https://www.

nist.gov/sites/default/files/documents/pml/wmd/metric/SP1038.pdf. For

more details, see http://www.amamanualofstyle.com/oso/public/jama/

si_conversion_table.html.

Abbreviations and Symbols

Use only standard abbreviations. Avoid abbreviations in the title and

abstract. The full term for an abbreviation should precede its first use

in the text, unless it is a standard abbreviation. All acronyms used in the

text should be expanded at first mention, followed by the abbreviation

in parentheses; thereafter the acronym only should appear in the text.

Acronyms may be used in the abstract if they occur 3 or more times

therein, but must be reintroduced in the body of the text. Generally,

abbreviations should be limited to those defined in the AMA Manual of

Style, current edition. A list of each abbreviation (and the corresponding

full term) used in the manuscript must be provided on the title page.

Online Manuscript Submission Process

The Turkish Journal of Hematology uses submission software powered

by ScholarOne Manuscripts. The website for submissions to The Turkish

Journal of Hematology is http://mc.manuscriptcentral.com/tjh. This

system is quick and convenient, both for authors and reviewers.

Setting Up an Account

New users to the submission site will need to register and enter their

account details before they can submit a manuscript. Log in, or click

the “Create Account” button if you are a first-time user. To create a new

account: After clicking the “Create Account” button, enter your name and

e-mail address, and then click the “Next” button. Your e-mail address is very

important. Enter your institution and address information, as appropriate,

and then click the “Next” Button. Enter a user ID and password of your

choice, select your area of expertise, and then click the “Finish” button.

A-IX


If you have an account, but have forgotten your log-in details, go to

“Password Help” on the journal’s online submission system and enter

your e-mail address. The system will send you an automatic user ID and

a new temporary password.

Full instructions and support are available on the site, and a user ID

and password can be obtained during your first visit. Full support for

authors is provided. Each page has a “Get Help Now” icon that connects

directly to the online support system. Contact the journal administrator

with any questions about submitting your manuscript to the journal

(tjh@tjh.com.tr). For ScholarOne Manuscripts customer support, click on

the “Get Help Now” link on the top right-hand corner of every page on

the site.

The Electronic Submission Process

Log in to your author center. Once you have logged in, click the “Submit

a Manuscript” link in the menu bar. Enter the appropriate data and

answer the questions. You may copy and paste directly from your

manuscript. Click the “Next” button on each screen to save your work

and advance to the next screen.

Upload Files

Click on the “Browse” button and locate the file on your computer. Select

the appropriate designation for each file in the drop-down menu next to

the “Browse” button. When you have selected all the files you want to

upload, click the “Upload Files” button. Review your submission before

sending to the journal. Click the “Submit” button when you are finished

reviewing. You can use ScholarOne Manuscripts at any time to check

the status of your submission. The journal’s editorial office will inform

you by e-mail once a decision has been made. After your manuscript

has been submitted, a checklist will then be completed by the Editorial

Assistant. The Editorial Assistant will check that the manuscript contains

all required components and adheres to the author guidelines. Once the

Editorial Assistant is satisfied with the manuscript it will be forwarded to

the Senior Editor, who will assign an editor and reviewers.

The Review Processs

Each manuscript submitted to The Turkish Journal of Hematology is

subject to an initial review by the editorial office in order to determine

if it is aligned with the journal’s aims and scope and complies with

essential requirements. Manuscripts sent for peer review will be assigned

to one of the journal’s associate editors that has expertise relevant to the

manuscript’s content. All accepted manuscripts are sent to a statistical

and English language editor before publishing. Once papers have been

reviewed, the reviewers’ comments are sent to the Editor, who will then

make a preliminary decision on the paper. At this stage, based on the

feedback from reviewers, manuscripts can be accepted or rejected, or

revisions can be recommended. Following initial peer-review, articles

judged worthy of further consideration often require revision. Revised

manuscripts generally must be received within 3 months of the date of

the initial decision. Extensions must be requested from the Associate

Editor at least 2 weeks before the 3-month revision deadline expires;

The Turkish Journal of Hematology will reject manuscripts that are

not received within the 3-month revision deadline. Manuscripts with

extensive revision recommendations will be sent for further review

(usually by the same reviewers) upon their re-submission. When a

manuscript is finally accepted for publication, the Technical Editor

undertakes a final edit and a marked-up copy will be e-mailed to the

corresponding author for review and any final adjustments.

Submission of Revised Papers

When revising a manuscript based on the reviewers’ and Editor’s

feedback, please insert all changed text in red. Please do not use

track changes, as this feature can make reading difficult. To submit

revised manuscripts, please log in to your author center at ScholarOne

Manuscripts. Your manuscript will be stored under “Manuscripts with

Decisions”. Please click on the “Create a Revision” link located to the right

of the manuscript title. A revised manuscript number will be created for

you; you will then need to click on the “Continue Submission” button.

You will then be guided through a submission process very similar to

that for new manuscripts. You will be able to amend any details you

wish. At stage 6 (“File Upload”), please delete the file for your original

manuscript and upload the revised version. Additionally, please upload

an anonymous cover letter, preferably in table format, including a

point-by-point response to the reviews’ revision recommendations. You

will then need to review your paper as a PDF and click the “Submit”

button. Your revised manuscript will have the same ID number as the

original version, but with the addition of an R and a number at the

end, for example, TJH-2011-0001 for an original and TJH-2011-0001.

R1, indicating a first revision; subsequent revisions will end with R2,

R3, and so on. Please do not submit a revised manuscript as a new

paper, as revised manuscripts are processed differently. If you click on

the “Create a Revision” button and receive a message stating that the

revision option has expired, please contact the Editorial Assistant at

tjh@tjh.com.tr to reactivate the option.

English Language and Statistical Editing

All manuscripts are professionally edited by an English language editor

prior to publication. After papers have been accepted for publication,

manuscript files are forwarded to the statistical and English language

editors before publishing. Editors will make changes to the manuscript

to ensure it adheres to TJH requirements. Significant changes or

concerns are referred to corresponding authors for editing.

Online Early

The Turkish Journal of Hematology publishes abstracts of accepted

manuscripts online in advance of their publication. Once an accepted

manuscript has been edited, the authors have submitted any final

corrections, and all changes have been incorporated, the manuscript will

be published online. At that time the manuscript will receive a Digital

Object Identifier (DOI) number. Both forms can be found at www.tjh.

com.tr. Authors of accepted manuscripts will receive electronic page

proofs directly from the printer and are responsible for proofreading

and checking the entire manuscript, including tables, figures, and

references. Page proofs must be returned within 48 hours to avoid delays

in publication.

A-X


CONTENTS

Research Articles

84 Comprehensive Mutation Profile in Acute Myeloid Leukemia Patients with RUNX1-RUNX1T1 or CBFB-MYH11

Fusions

Wei Qin, Xiayu Chen, Hong Jie Shen, Zheng Wang, Xiaohui Cai, Naike Jiang, Haiying Hua; Changzhou, Wuxi, Soochow, Suzhou, China

94 Invasive Fungal Infections in Children with Leukemia: Clinical Features and Prognosis

Melike Sezgin Evim, Özlem Tüfekçi, Birol Baytan, Hale Ören, Solmaz Çelebi, Beyza Ener, Kevser Üstün Elmas, Şebnem Yılmaz, Melek

Erdem, Mustafa Kemal Hacımustafaoğlu, Adalet Meral Güneş; Bursa, İzmir, Turkey

103 Eltrombopag for Treatment of Thrombocytopenia Following Hematopoietic Stem Cell Transplantation

Zeynep Tuğba Güven, Serhat Çelik, Bülent Eser, Mustafa Çetin, Ali Ünal, Leylagül Kaynar; Kayseri, Antalya, Turkey

109 Assessment of Bone Marrow Biopsy and Cytogenetic Findings in Patients with Multiple Myeloma

Ahmet Şeyhanlı, Boran Yavuz, Zehra Akşit, Zeynep Yüce, Sermin Özkal, Oğuz Altungöz, Fatih Demirkan, İnci Alacacıoğlu,

Güner Hayri Özsan; Sivas, İzmir, Turkey

117 Combination of Haploidentical Hematopoietic Stem Cell Transplantation with Umbilical Cord-Derived

Mesenchymal Stem Cells in Patients with Severe Aplastic Anemia: A Retrospective Controlled Study

Xian-Fu Sheng, Hui Li, Li-Li Hong, Hai-Feng Zhuang; Zhejiang, China

130 Castleman Disease: A Multicenter Case Series from Turkey

Eren Gündüz, Hakkı Onur Kırkızlar, Elif Gülsüm Ümit, Sedanur Karaman Gülsaran, Vildan Özkocaman, Fahir Özkalemkaş,

Ömer Candar, Tugrul Elverdi, Selin Küçükyurt, Semra Paydaş, Özcan Çeneli, Sema Karakuş, Senem Maral, Ömer Ekinci, Yıldız İpek,

Cem Kis, Zeynep Tuğba Güven, Aydan Akdeniz, Tiraje Celkan, Ayşe Hilal Eroğlu Küçükdiler, Gülsüm Akgün Çağlıyan, Ceyda Özçelik

Şengöz, Ayşe Karataş, Tuba Bulduk, Alper Özcan, Fatma Burcu Belen Apak, Aylin Canbolat, İbrahim Kartal, Hale Ören, Ersin Töret,

Gül Nihal Özdemir, Şule Mine Bakanay Öztürk; Eskişehir, Edirne, Bursa, İstanbul, Adana, Konya, Ankara, Elazığ, Kayseri, Mersin,

Aydın, Denizli, Trabzon, Samsun, İzmir, Turkey

Perspective in Hematology

136 Gaucher Disease for Hematologists

Gül Nihal Özdemir, Eren Gündüz; İstanbul, Eskişehir, Turkey

Images in Hematology

140 Gallbladder Involvement of Diffuse Large B-Cell Lymphoma with 18 F-FDG PET/CT

Esra Arslan, Göksel Alçin, Tamer Aksoy, Tevfik Fikret Çermik; İstanbul, Turkey

142 Mast Cell Leukemia in a Patient with Teratoma

Yan Shen, Zhenni Wang, Huijun Lin, Jinlin Liu; Hangzhou, China

A-XI


Letters to the Editor

144 Caution Regarding the Difference Between Flower-Like Lymphocytes and Flower-Like Plasma Cells

Jingnan Zhu, Zhang Li, Yong Wang, Jinlin Liu; Hangzhou, Shaoxing, Ningbo, China

146 Does Treatment of Hepatitis C Reduce Inhibitor Titers in Hemophilia A?

Memiş Hilmi Atay, Emine Türkoğlu, Engin Kelkitli; Samsun, Tokat, Turkey

148 Spontaneous Remission in Paroxysmal Nocturnal Hemoglobinuria: An Extremely Rare Case

Özgür Mehtap, Ayfer Gedük; Kocaeli, Turkey

150 Hematopoietic Stem Cell Transplantation to a Patient with Acute Myeloid Leukemia from a Sibling Donor

Positive for SARS-CoV-2 by RT-PCR Test

Ahmet Koç, Ömer Doğru, Nurşah Eker, Burcu Tufan Taş, Rabia Emel Şenay; İstanbul, Turkey

A-XII


RESEARCH ARTICLE

DOI: 10.4274/tjh.galenos.2022.2021.0641

Turk J Hematol 2022;39:84-93

Comprehensive Mutation Profile in Acute Myeloid Leukemia

Patients with RUNX1-RUNX1T1 or CBFB-MYH11 Fusions

RUNX1-RUNX1T1 veya CBFB-MYH11 Füzyonları Olan Akut Myeloid Lösemili Hastalarda

Detaylı Mutasyon Profili

Wei Qin 1 *, Xiayu Chen 2 *, Hong Jie Shen 3 , Zheng Wang 3,4 , Xiaohui Cai 1 , Naike Jiang 1 , Haiying Hua 2

1Affiliated Changzhou Second Hospital of Nanjing Medical University, Department of Hematology, Changzhou, China

2Affiliated Hospital of Jiangnan University, Department of Hematology, Wuxi, China

3The First Affiliated Hospital of Soochow University, Department of Hematology, Soochow, China

4Suzhou Jsuniwell Medical Laboratory, Suzhou, China

*These authors contributed equally to this work.

Abstract

Objective: This study was undertaken with the aim of better

understanding the genomic landscape of core-binding factor (CBF)

acute myeloid leukemia (AML).

Materials and Methods: We retrospectively analyzed 112 genes that

were detected using next-generation sequencing in 134 patients

with de novo CBF-AML. FLT3-ITD, NPM1, and CEBPA mutations were

detected by DNA-PCR and Sanger sequencing.

Results: In the whole cohort, the most commonly mutated

genes were c-KIT (33.6%) and NRAS (33.6%), followed by FLT3

(18.7%), KRAS (13.4%), RELN (8.2%), and NOTCH1 (8.2%). The

frequencies of mutated genes associated with epigenetic

modification, such as IDH1, IDH2, DNMT3A, and TET2, were low,

being present in 1.5%, 0.7%, 2.2%, and 7.5% of the total

number of patients, respectively. Inv(16)/t(16;16) AML patients

exhibited more mutations of NRAS and KRAS (p=0.001

and 0.0001, respectively) than t(8;21) AML patients. Functionally

mutated genes involved in signaling pathways were observed more

frequently in the inv(16)/t(16;16) AML group (p=0.016), while the

mutations involved in cohesin were found more frequently in

the t(8;21) AML group (p=0.011). Significantly higher white blood cell

counts were found in inv(16)/t(16;16) AML patients with c-KIT (c-KIT mut )

or NRAS (NRAS mut ) mutations compared to the corresponding t(8;21)

AML/c-KIT mut and t(8;21) AML/NRAS mut groups (p=0.001 and 0.009,

respectively).

Conclusion: The mutation profiles of t(8;21) AML patients showed

evident differences from those of patients with inv(16)/t(16;16)

AML. We have provided a comprehensive overview of the mutational

landscape of CBF-AML.

Keywords: Core-binding factor, Acute myeloid leukemia, Mutation,

Next-generation sequencing

Öz

Amaç: Bu çalışma çekirdek bağlama faktörü (ÇBF) akut myeloid

löseminin (AML) genomik durumunu daha iyi anlamak amacıyla

yapılmıştır.

Yöntemler: Yüz otuz dört de novo ÇBF-AML hastasında yeni nesil

dizileme ile tespit edilen 112 geni geriye dönük olarak analiz ettik.

FLT3-ITD, NPM1 ve CEBPA mutasyonları DNA-PCR ve Sanger dizileme

ile tespit edildi.

Bulgular: Bütün kohortta en sık mutasyonlu genler c-KIT (33,6%) ve

NRAS (33,6%) ve ardından FLT3 (%18,7), KRAS (%13,4), RELN (%8,2),

NOTCH1 (%8,2) idi. IDH1, IDH2, DNMT3A ve TET2 gibi epigenetik

modifikasyonla ilişkili mutasyona uğramış genlerin sıklığı düşüktü ve

toplam hastaların sırasıyla %1,5, %0,7, %2,2 ve %7,5’inde mevcuttu.

Inv(16)/t(16;16) AML hastalarında NRAS ve KRAS mutasyonları

t(8;21) AML hastalarına göre daha fazlaydı (sırasıyla; p=0,001;

0,0001). İşlevsel olarak sinyal yolaklarında yer alan mutasyonlu genler

inv(16)/t(16;16) AML grubunda daha çok gözlenirken (p=0,016),

kohezin içinde yer alan mutasyonlar t(8;21) AML grubunda daha

çok bulundu (p=0,011). c-KIT (c-KIT mut ) veya NRAS mutasyonları

(NRAS mut ) olan inv(16)/t(16;16) AML hastalarında karşılığındaki t(8;21)

AML/c-KIT mut ve t(8;21) AML/NRAS mut gruplarına göre beyaz küre sayısı

daha yüksek bulundu (sırasıyla; p=0,001; 0,009,).

Sonuç: t(8;21) AML hastalarının mutasyon profilleri inv(16)/t(16;16)

AML’den belirgin farklılıklar gösterdi. Bu çalışmada ÇBF-AML’nin

mutasyon profili kapsamlı bir biçimde incelenmiştir.

Anahtar Sözcükler: Çekirdek bağlama faktörü, Akut myeloid lösemi,

Mutasyon, Yeni nesil sekanslama

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Naike Jiang, M.D., Affiliated Changzhou Second Hospital of

Nanjing Medical University, Department of Hematology, Changzhou China

Phone : 8613357894897

E-mail : naike.j@163.com ORCID: orcid.org/0000-0003-4495-567X

Received/Geliş tarihi: November 20, 2021

Accepted/Kabul tarihi: April 21, 2022

84


Turk J Hematol 2022;39:84-93

Qin W. et al: Mutation Profile in CBF-AML

Introduction

Cases of acute myeloid leukemia (AML) involving the

core-binding factor (CBF) include AML with t(8;21) and

inv(16)/t(16;16) chromosomal translocations, leading to

the RUNX-RUNX1T1 and CBFB-MYH11 fusions genes,

respectively. Such AML patients account for approximately

25% of pediatric and 15% of adult de novo AML cases [1],

and CBF-AML was recognized as a unique entity in the

2016 World Health Organization classification of myeloid

neoplasms and acute leukemia [2]. Accumulating evidence

has revealed that the t(8;21)(q22;q22) and inv(16)/t(16;16) CBF

rearrangements are associated with favorable outcomes relative

to other cytogenetic subtypes and that allogeneic

hematopoietic stem cell transplantation is not generally

recommended during the first complete remission (CR)

[3,4]. However, relapse occurs in up to 40% of these cases,

indicating clinicopathological heterogeneity within this

AML subset [5,6,7,8]. Further investigation is still needed to

better understand leukemogenesis and disease progression.

Previous findings have demonstrated that expressions of

translocation-encoded AML1 or CBF fusion proteins are

insufficient by themselves to induce a full leukemic phenotype

[9]. Further evidence supporting this model comes from

the fact that mutations in genes activating tyrosine kinase

signaling (including KIT, N/KRAS, and FLT3) are frequent in

both CBF-AML subtypes [6,10]. Nonetheless, data regarding

the prognostic significance of KIT and RAS in CBF-AML

are contradictory. Duployez et al. [11] reported the presence of

additional aberrations in >90% of CBF-AML cases, and mutations

in epigenetic modifications or cohesin genes were associated

with poor prognosis in t(8;21) AML patients with TK pathway

mutations using next-generation sequencing (NGS). Ishikawa

et al. [12] revealed that the c-KIT exon 17 mutation and the

presence of extramedullary tumors in t(8;21) AML patients were

poor prognostic factors for relapse-free survival, as were the loss

of chromosome X or Y and NRAS mutation in patients with

inv(16)/t(16;16). These findings highlight the multiclonality

of CBF-AML and suggest that the prognostic impact may

differ in the context of certain gene mutations between AML

patients with t(8;21) and those with inv(16)/t(16;16).

Comprehensive genetic analysis using NGS may be helpful in

refining our understanding of the prognosis of CBF-AML [11]. To

the best of our knowledge, limited data are available regarding

the impact of companion gene mutations in CBF-AML. To better

characterize this subtype and to better understand the role of

co-mutations in CBF-AML, we performed extensive mutational

analysis by NGS for 134 CBF-AML patients. The clinical value of

co-mutations in CBF-AML patients was also explored.

Materials and Methods

Patients

A total of 134 newly diagnosed de novo CBF-AML patients

were selected from the Affiliated Changzhou Second Hospital

of Nanjing Medical University, Wuxi Third People’s Hospital, and

First Affiliated Hospital of Soochow University from May

2016 to June 2021. The diagnosis of CBF-AML was based on the

2008 definition of the World Health Organization [2]. Eighty AML

patients with t(8;21)/RUNX1-RUNX1T1 and 54 patients

with inv(16)/t(16;16)/CBFB-MYH11 were included in the

analysis. The study was approved by the local ethics committee

and conducted in accordance with the Declaration of Helsinki.

Mutational Analysis by Next-Generation Sequencing

Genomic DNA was extracted from fresh bone marrow or

peripheral blood samples with the QIAamp DNA Mini Kit

(QIAGEN GmbH, Germany) following the manufacturer’s

instructions. The NGS library was prepared using at least 200 ng

of genomic DNA. Massively parallel sequencing was performed

with an Illumina next-generation sequencer and a variant allele

frequency of >3% was used as the threshold for calling singlenucleotide

variants. A high depth of coverage (1000x) was

obtained for 112 genes, including whole coding regions known

to be frequently mutated in hematological malignancies, such

as genes involved in epigenetic regulators, signaling pathways,

transcription factors, spliceosomes, cohesin complex, tumor

suppressors, and chromatin modifiers (Table 1). Searches were

performed in the COSMIC database for altered DNA sequences

deemed to be mutations or variants with IGV software and

were confirmed in the SNP database (dbSNP). Polymerase chain

reaction (PCR) followed by direct Sanger sequencing was used

to detect FLT3-ITD, NPM1 (exon 12), and CEBPA to avoid possible

false-negative results due to limitations of NGS, as previously

described [13,14].

Other Cytogenetic and Molecular Abnormality Screening

Bone marrow (BM) cells were collected and cultured at the

time of the initial diagnosis. The presence of the t(8;21)

or inv(16)/t(16;16) rearrangement was determined by the

conventional G/R banding method. Fluorescence in situ

hybridization (FISH) of interphase nuclei and/or metaphases was

performed for the chimeric genes RUNX1-RUNX1T1 and CBFB-

MYH11. The fusion transcripts were identified by real-time

quantitative PCR (RT-qPCR) method using bone marrow

or peripheral blood samples at diagnosis, as previously described

[15].

Statistical Analysis

CR was defined as <5% blast cells, no Auer rods, and no clusters

of blast cells by bone marrow analysis as well as no evidence

85


Qin W. et al: Mutation Profile in CBF-AML

Turk J Hematol 2022;39:84-93

of extramedullary leukemia. All statistical analyses of the data

were carried out using IBM SPSS Statistics 20.0 (IBM Corp.,

Armonk, NY, USA). The chi-square and Fisher exact tests were

used for comparisons of categorical variables between different

cohorts. The Student t-test was used to analyze continuous

variables with normal distribution and the Mann-Whitney U test

was used for data that did not comply with normal

distribution. Values of p<0.05 were considered statistically

significant.

Results

Patient Characteristics

The 134 CBF-AML patients enrolled in this study

included 65 women and 69 men with a median age of 35.5

years (range: 16.0-73.0 years). The median white blood cell

(WBC) count was 15.3x10 9 /L (range: 0.9 to 156.0x10 9 /L),

median hemoglobin (Hb) level was 81 g/L (range: 39.0

to 124.0 g/L), and median platelet count was 28.0x10 9/ L

(range: 2.0 to 170.0x10 9 /L). Patients with inv(16) or

t(16;16) AML tended to be older than t(8;21) AML patients

(41 vs. 32 years, p=0.048) and also had higher WBC counts

(35.9x10 9 /L vs. 7.8x10 9 /L, p=0.0001). No significant differences

were identified regarding gender, Hb level, or platelet

count between patients with t(8;21) and inv(16)/t(16;16) AML.

By conventional chromosome analysis, 41.0% (55/134) of the

CBF-AML patients were found to have secondary cytogenetic

abnormalities. Additional chromosomal alterations were found

in 45.0% (36/80) and 35.2% (19/54) of the patients with

t(8;21) and inv(16)/t(16;16), respectively. Loss of the X or Y

chromosome was identified as the most common secondary

alteration (33/134, 24.6%), followed by trisomy 22 (13/134,

9.7%). In patients with inv(16)/t(16;16), the most frequently

identified additional cytogenetic alterations were trisomy 22

and trisomy 8, which were found in 20.1% (13/54) and 9.3%

(5/54) of these patients, respectively.

Table 1. The 112 genes analyzed in this study.

Number Gene Number Gene Number Gene Number Gene

1 ABL1 29 DNM2 57 KMT2A 85 SETBP1

2 ACD 30 DNMT3A 58 KRAS 86 SETD2

3 ANKRD26 31 DNMT3B 59 MAPK1 87 SF1

4 ARIDIA 32 ECT2L 60 KMT2D 88 SF3A1

5 ASXL1 33 EED 61 MPL 89 SF3B1

6 ATG2B 34 EP300 62 MYC 90 SH2B3

7 ATM 35 ETNK1 63 MYD88 91 SMC1A

8 B2M 36 EZH2 64 NF1 92 SMC3

9 BCOR 37 FAM46C 65 NOTCH1 93 SRP72

10 BCOR1 38 BRINP3 66 NOTCH2 94 SRSF2

11 BIRC3 39 FAT1 67 NPM1 95 STAG2

12 BRAF 40 FBXW7 68 NRAS 96 STAT3

13 CALR 41 FGFR3 69 PAX5 97 SUZ12

14 CBL 42 FLT3 70 PDS5B 98 ETV6

15 CCND1 43 GATA1 71 PHF6 99 TERC

16 CCND3 44 GATA2 72 PIGA 100 TERT

17 CCR4 45 GATA3 73 PLCG1 101 TET2

18 CD79B 46 JAK1 74 PRKCB 102 TNFAIP3

19 CDC25C 47 JAK2 75 PRPF40B 103 TP53

20 CDKN2A 48 JAK3 76 PRPS1 104 TPMT

21 CEBPA 49 HNRNPK 77 PTEN 105 TRAF3

22 CREBBP 50 ID3 78 PTPN11 106 U2AF1

23 CSF3R 51 IDH1 79 RAD21 107 U2AF2

24 CUX1 52 IDH2 80 RB1 108 WHSC1

25 CXCR4 53 IKZF1 81 RBBP6 109 WT1

26 DDX3X 54 IL7R 82 RELN 110 XPO1

27 DDX41 55 KDM6A 83 RHOA 111 ZRSR2

28 DIS3 56 KIT 84 RUNX1 112 ZMYM3

86


Turk J Hematol 2022;39:84-93

Qin W. et al: Mutation Profile in CBF-AML

Trisomies 8 and 22 were more frequently observed in

inv(16)/t(16;16) AML patients (p=0.006 and 0.0001, respectively),

while loss of the X or Y chromosome was more common in

patients with t(8;21) AML (p=0.0001). Del(9q) was not noted in

any inv(16) patients, while trisomies 8 and 22 were not found in

any t(8;21)patients. In all cases for which FISH and/or RT-qPCR

testing was performed, the results were in agreement with the

results of chromosome analysis (data not shown). Both CD19

and CD56 antigen expressions were observed more frequently in

the t(8;21) AML group (both p=0.0001). Clinical and biological

characteristics of the patients are provided in Table 2.

Comparison of Clinical Features and Incidence of

Genetic Mutations Between AML Patients with t(8;21)

and inv(16)/t(16;16)

Among the participating 134 CBF-AML patients, 68 mutated

genes were detected by screening the 112-gene panel. Thirtytwo

of those 68 genes could be classified as transcription

factor, DNA methylation, signaling, spliceosome, cohesin,

or tumor suppressor genes. An average of 3.19 (range: 1-10)

mutations per individual were detected among these CBF-

AML cases. While 22 patients had 1 alteration, 20 had 2, 32 had

3, and 60 had 4 or more. Among all genes sequenced, the most

commonly mutated genes were c-KIT (45/134, 33.6%) and NRAS

(45/134, 33.6%), followed by FLT3 (25/134, 18.7%), KRAS

(18/134, 13.4%), RELN (11/134, 8.2%), NOTCH1 (11/134,

8.2%), TET2 (10/134, 7.5%), and WT1 (10/134, 7.5%). The other

genes had mutation prevalences of <5%. The most frequently

affected functional pathway was the signaling pathway, with

such mutations observed in as many as 86.6% of cases. In

addition, our findings suggest that the frequencies of

mutations in genes associated with epigenetic modification,

such as IDH1, IDH2, DNMT3A, and TET2, are low in

CBF-AML, being identified in 1.5%, 0.7%, 2.2%, and 7.5% of the

total number of participating patients, respectively.

Concomitant gene abnormalities were found in 100% of

the patients with t(8;21) and 100% of the patients with

inv(16)/t(16;16). The patients with inv(16)/t(16;16) AML were

found to have more mutations in the NRAS and KRAS genes

(53.7% vs. 20.0%, p=0.001 and 27.8% vs. 3.8%, p=0.0001,

respectively) compared to t(8;21) AML patients. The distributions

of c-KIT, FLT3, RELN, TET2, FAT1, and NOTCH1 mutations

within these two groups were similar (Table 3). Functionally

mutated genes involved in signaling pathways were

observed more frequently in the inv(16)/t(16;16) AML group

Table 2. Clinical and biological characteristics of the CBF-AML patients at diagnosis.

Variable

Gender

Total

(n=134)

RUNX1-RUNX1T1

(n=80)

CBFB-MYH11

(n=54)

Male, n (%) 69 (51.5%) 42 (52.5%) 27 (50%)

Female, n (%) 65 (48.5%) 38 (47.5%) 27 (50%)

Age (years)

Median (range) 35.5 (16-73) 32 (16-73) 41 (16-63) 0.048

WBC count (x10 9 /L)

Median (range) 15.3 (0.9-156.0) 7.8 (0.9-123) 35.9 (1.6-156) 0.0001

Hb (g/L)

Median (range) 81 (39.0-124.0) 78 (39-120) 86.5 (40-124) 0.169

PLT count (x10 9 /L)

Median (range) 28.0 (2.0-170.0) 32 (2-170) 26 (3-121) 0.725

Secondary cytogenetic abnormalities

Loss of X or Y chromosome, n (%) 33 (24.6%) 32 (40.0%) 1 (1.9%) 0.0001

del(9q), n (%) 4 (3.0%) 4 (5.0%) 0 0.095

Trisomy 8, n (%) 5 (3.7%) 0 5 (9.3%) 0.006

Trisomy 22, n (%) 13 (9.7%) 0 13 (20.1%) 0.0001

Immunophenotyping

CD19 expression, n (n/N, %) 59 (59/116, 50.86%) 58 (58/73, 79.5%) 1 (1/43, 2.33%) 0.0001

NA, n 18 7 11

CD56 expression, n (n/N, %) 51 (51/92, 55.43%) 50 (50/58, 86.21%) 1 (1/34, 2.94%) 0.0001

NA, n 42 22 20

CBF: Core-binding factor; AML: acute myeloid leukemia; WBC: white blood cell; Hb: hemoglobin; PLT: platelet; NA: not available.

p

0.776

87


Qin W. et al: Mutation Profile in CBF-AML

Turk J Hematol 2022;39:84-93

(p=0.016), while cohesin mutations were found more frequently

in the t(8;21) AML group (11.3% vs. 0%, p=0.011). All cohesin

mutations were mutually exclusive among each other

(Figure 1). The mutation distribution is provided in detail

in Table 3.

Relationships Between Clinical Characteristics, CR

Rate, and Mutations

We analyzed the clinical characteristics of patients with

mutations in c-KIT, NRAS, KRAS, CSF3R, TET2, and FLT3-ITD. As

listed in Table 4, significantly higher WBC counts were found

in inv(16)/t(16;16) AML patients with c-KIT (c-KIT mut ) or NRAS

(NRAS mut ) mutations than in t(8;21) AML patients with

c-KIT mut and NRAS mut (p=0.001 and 0.009, respectively).

Patients with both inv(16)/t(16;16) AML/TET2 mut and

inv(16)/t(16;16) AML/FLT3-ITD mut also had higher WBC

counts than those with t(8;21) AML/TET2 mut and t(8;21)

AML/FLT3-ITD mut , but these differences did not reach statistical

significance (p=0.088 and 0.067, respectively). No difference

was found between other factors such as age, gender, Hb

level, or platelet count.

This study also aimed to assess the impact of common gene

mutations on the rate of CR after initial induction

therapy. Among the 134 participating patients, relevant data

were available for 128. The overall CR rate among these cases

was 94.53% (121/128). No differences in CR rate were observed

according to mutated genes (KIT, NRAS, KRAS, TET2, FLT3-ITD)

between t(8;21) AML and inv(16)/t(16;16) AML patients. Clinical

characteristics and CR rates of CBF-AML patients with common

mutations are shown in Table 4.

Discussion

In this study, patients with inv(16)/t(16;16) AML had higher WBC

counts than those with t(8;21) AML. Trisomies 8 and 22 were

more frequently observed in inv(16) patients, while loss of the

X or Y chromosome was more common in t(8;21) AML. Patients

with t(8;21) AML also expressed CD19 and CD56 more frequently

than those with inv(16) AML. These findings are consistent with

the conclusions of previous reports [11,16].

Figure 1. Comparisons of genetic mutations between AML patients with t(8;21) and inv(16)/t(16;16).

AML: Acute myleoid leukemia

88


Turk J Hematol 2022;39:84-93

Qin W. et al: Mutation Profile in CBF-AML

Table 3. Concomitant gene abnormalities of CBF-AML at diagnosis.

Total

Mutational genes

(n=134)

RUNX1-RUNX1T1

(n=80)

CBFB-MYH11

(n=54)

NPM1 3 (2.2%) 2 (2.5%) 1 (1.9%) 0.804

Signaling pathways, n (%) 116 (86.6%) 64 (80.0%) 52 (96.3%) 0.016

c-KIT, n (%) 45 (33.6%) 28 (35.0%) 17 (31.5%) 0.672

NRAS, n (%) 45 (33.6%) 16 (20.0%) 29 (53.7%) 0.0001

KRAS, n (%) 18 (13.4%) 3 (3.8%) 15 (27.8%) 0.0001

FLT3, n (%) 25 (18.7%) 15 (18.8%) 10 (18.5%) 1

CSF3R, n (%) 8 (6.0%) 7 (8.8%) 1 (1.9%) 0.143

RELN, n (%) 11 (8.2%) 8 (10.0%) 3 (5.6%) 0.524

NOTCH1, n (%) 11 (8.2%) 7 (8.8%) 4 (7.4%) 1

NOTCH2, n (%) 8 (6.0%) 4 (5.0%) 4 (7.4%) 0.714

JAK2, n (%) 8 (6.0%) 6 (7.5%) 2 (3.7%) 0.363

SH2B3, n (%) 6 (4.5%) 6 (7.5%) 0 0.081

PTPN11, n (%) 3 (2.2%) 1 (1.3%) 2 (3.7%) 0.565

Epigenetic regulators, n (%) 15 (11.2%) 10 (12.5%) 5 (9.3%) 0.781

TET2, n (%) 10 (7.5%) 6 (7.5%) 4 (7.4%) 1

IDH1, n (%) 2 (1.5%) 1 (1.3%) 1 (1.9%) 1

IDH2, n (%) 1 (0.7%) 1 (1.3%) 0 1

DNMT3A, n (%) 3 (2.2%) 3 (3.8%) 0 0.273

Transcription factors, n (%) 17 (12.7%) 8 (10.0%) 9 (16.7%) 0.296

ETV6, n (%) 1 (0.7%) 1 (1.3%) 0 1

RUNX1, n (%) 2 (1.5%) 1 (1.3%) 1 (1.9%) 1

GATA2, n (%) 1 (0.7%) 1 (1.3%) 0 1

SETBP1, n (%) 8 (6.0%) 3 (3.8%) 5 (9.3%) 0.267

CEBPA dm , n (%) 7 (5.2%) 3 (3.8%) 4 (7.4%) 0.439

Spliceosomes, n (%) 4 (3.0%) 4 (5.0%) 0 0.148

SRSF2, n (%) 1 (0.7%) 1 (1.3%) 0 1

SF3B1, n (%) 3 (2.2%) 3 (3.8%) 0 0.273

Tumor suppressors, n (%) 14 (10.45%) 9 (11.25%) 5 (9.26%) 0.712

TP53, n (%) 4 (3.0%) 4 (5.0%) 0 0.148

WT1, n (%) 10 (7.5%) 5 (6.3%) 5 (9.3%) 0.516

Cohesin, n (%) 9 (6.7%) 9 (11.3%) 0 0.011

RAD21, n (%) 4 (3.0%) 4 (5.0%) 0 0.148

SMC1A, n (%) 3 (2.2%) 3 (3.8%) 0 0.273

SMC3, n (%) 2 (1.5%) 2 (2.5%) 0 0.515

Chromatin modifiers, n (%) 15 (11.2%) 11 (13.8%) 4 (7.4%) 0.403

ASXL1, n (%) 8 (6.0%) 6 (7.5%) 3 (5.6%) 0.659

KDM6A, n (%) 2 (1.5%) 2 (2.5%) 0 0.515

BCOR, n (%) 4 (3.0%) 3 (3.8%) 1 (1.9%) 0.648

BCORL1, n (%) 1 (0.7%) 1 (1.3%) 0 1

Number of mutated genes

1 22 (16.4%) 14 (17.5%) 8 (14.8%) 0.681

2 20 (14.9%) 10 (12.5%) 10 (18.5%) 0.338

3 32 (23.9%) 18 (22.5%) 14 (25.9%) 0.648

≥4 60 (44.8%) 38 (47.5%) 22 (40.7%) 0.440

Average number (range) 3.19 (1-10) 3.17 (1-8) 3.22 (1-10) 0.873

CBF: Core-binding factor; AML: acute myeloid leukemia.

p

89


Qin W. et al: Mutation Profile in CBF-AML

Turk J Hematol 2022;39:84-93

Table 4. Clinical characteristics of the common co-mutations in patients with RUNX1-RUNX1T1 or CBFB-MYH11 fusions.

Variables

Age, years,

median (range)

Male/female,

n/n

WBC count,

x10 9 /L, median

(range)

Hb, g/L,

median

(range)

PLT count,

x10 9 /L, median

(range)

CR rate, n/N

(%)

c-KIT mutations (n=45)

RUNX1-RUNX1T1 (n=28) 31.5 (16-56) 13/15 10.1 (2.0-49.9) 78 (38-19) 32.5 (11-170) 50.0% (13/26) *

CBFB-MYH11 (n=17) 32 (16-57) 10/7 29 (5.7-137) 93 (63-124) 40 (15-117) 58.8% (10/17)

p 0.566 0.542 0.001 0.081 0.419 0.571

NRAS mutations (n=45)

RUNX1-RUNX1T1 (n=16) 30.5 (16-69) 9/7 18.6 (2.4-123) 80 (57-119) 28 (7-105) 87.5% (14/16)

CBFB-MYH11 (n=29) 35 (18-63) 14/15 43 (5.0-156) 87 (40-122) 25 (3-117) 92.8% (26/28) *

p 0.618 0.758 0.009 0.506 0.585 0.552

KRAS mutations (n=18)

RUNX1-RUNX1T1 (n=3) 41 (16-51) 3/0 30.9 (3.2-49.9) 79 (64-110) 11 (7-58) 100% (3/3)

CBFB-MYH11 (n=15) 36 (19-58) 9/6 32 (1.56-144) 82 (59-110) 40 (15-121) 80% (12/15)

p 0.824 0.515 0.783 0.824 0.138 1.000

CSF3R mutations (n=8)

RUNX1-RUNX1T1 (n=7) 21 (16-32) 2/5 5.1 (0.9-52.4) 63 (59-119) 28 (8-105) 57.1% (4/7)

CBFB-MYH11 (n=1) 56 (56-56) 1/0 34.9 98 (98-98) 14 (14-14) 100% (1/1)

p 0.127 0.375 0.275 0.275 0.275 1.000

TET2 mutations (n=10)

RUNX1-RUNX1T1 (n=6) 36 (29-49) 2/4 4.2 (0.9-74) 89.5 (45-107) 30.5 (2-80) 66.7% (4/6)

CBFB-MYH11 (n=4) 39.5 (26-52) 1/3 28.6 (18.3-65.4) 77.5 (59-98) 22 (22-44) 100% (4/4)

p 1.00 1 0.088 0.593 0.892 0.467

FLT3-ITD mutations (n=10)

RUNX1-RUNX1T1 (n=7) 43 (24-64) 2/5 23.9 (5.6-40) 75 (57-108) 23 (6-45) 42.9% (3/7)

CBFB-MYH11 (n=3) 40 (36-42) 3/0 54 (33-137) 82 (77-85) 40 (20-57) 66.7% (2/3)

p 0.732 0.167 0.067 0.21 0.138 1.000

WBC: White blood cell; HB: hemoglobin; PLT: platelet; CR: complete remission. *: Some patients did not receive any chemotherapy or were only treated with low-dose cytosine

arabinoside.

Both t(8;21) and inv(16)/t(16;16) disrupt the normal functioning

of the heterodimeric transcription factor CBF complex in AML

with relatively similar clinical outcomes. However, the molecular

genetic abnormalities potentially explaining the differences

between these two AML subtypes have not yet been explored in

detail. We performed extensive mutational analysis by NGS for

134 patients with CBF-AML who ranged in age from 16 to 73

years. As expected, additional aberrations were found in 100%

of these CBF-AML cases and the most commonly mutated gene

was c-KIT, as seen in 35.0% of AML cases with t(8;21) and

31.5% of AML cases with inv(16)/t(16;16). This is in accordance

with the previous research conducted by Duployez et al.

[11]. Interestingly, a significantly different spectrum of gene

mutations was demonstrated in AML between patients with

90


Turk J Hematol 2022;39:84-93

Qin W. et al: Mutation Profile in CBF-AML

t(8;21) and inv(16)/t(16;16). We noticed that fewer signaling

pathways were involved in cases of t(8;21) in comparison

to inv(16)/t(16;16) (80.0% vs. 96.3%, p=0.016), while patients

with inv(16)/t(16;16) AML exhibited more mutations

in KRAS and NRAS compared to t(8;21) AML patients.

RAS genes encode a family of membrane-associated proteins that

regulate signal transduction upon the ligand binding to a variety

of membrane receptors, and they play important roles in physical

processes including proliferation, differentiation, and apoptosis

[17,18]. Activating point mutations of RAS genes have generally

been accepted as oncogenic events in the tumorigenesis of

numerous malignancies, including hematological malignancies

such as AML [11,19,20]. RAS mutations seem to be particularly

frequent in inv(16)/t(16;16) AML, with a reported incidence

of up to 54% [11]. Duployez et al. [11] and Boissel et

al. [21] reported that NRAS and KRAS mutations were more

common among AML patients with inv(16)/t(16;16) than those

with t(8;21). Further studies showed that RAS mutations had

no effect on overall/disease-free survival, CR, or relapse rates

[22,23,24]. In our cohort, both NRAS and KRAS mutations were

more frequently found among inv(16)/t(16;16)

AML patients than in the t(8;21) AML group, a finding previously

demonstrated among other cohorts [11,21]. These data suggest

that the synergic cooperation between inv(16)/t(16;16) and RAS

mutations may influence the pathophysiology of CBF-AML.

Cohesin is a multimeric protein complex that is involved

in the cohesion of sister chromatids, post-replicative DNA

repair, and transcriptional regulation and it is composed of 4

core subunits: the SMC1A, SMC3, RAD21, and STAG proteins

[25]. Cohesin mutations have been reported in about 6%

of AML patients [26] and fewer than 2% of cases of CBF-

AML [11]. Recent data revealed the identification of cohesin

and chromatin modifier mutations in t(8;21) but not inv(16)

patients [11,27]. In the present study, mutations in genes

encoding members of the cohesin complex were present in

11.3% of the t(8;21) AML patients and none of the inv(16) AML

patients, and all cohesin mutations were mutually exclusive

among each other, which is consistent with previous studies

[26,27]. It is interesting that cohesin gene mutations are more

frequent in patients with RUNX1-mutated AML [28]. Indeed,

Mazumdar et al. [29] demonstrated that cohesin mutations led

to a state of elevated chromatin accessibility and higher levels

of binding at RUNX1 binding sites. These findings suggest links

between alterations in cohesin and the RUNX1-RUNXT1 fusion

oncoprotein. In addition, our study also suggests that the

frequencies of mutations in genes associated with epigenetic

modification (IDH1, IDH2, DNMT3A, and TET2), are low in CBF-

AML, which is in accordance with the findings of Park et al.

[30] and Duployez et al. [11]. These results may support the

idea that mutations involved in epigenetic modification do not

contribute to leukemogenesis in CBF-AML.

Yang et al. [23] showed that AML patients with RAS mutations had

significantly higher WBC counts at diagnosis than those

without mutations (p=0.001). Boissel et al. [21] demonstrated

that CBF-AML patients with c-KIT mutations had a significantly

higher median WBC count at presentation, but this difference

was mainly observed among patients with inv(16), with

the mutations being less common in patients with t(8;21).

Additionally, no gene mutations predicted poor response to

induction in comparisons with patients without mutations for

particular genes (c-KIT, RAS), except for FLT3 [21]. Jahn et al.

[31] found that both DNMT3A and TET2 were associated with

a significantly worse prognosis in univariate analysis. However,

regarding the subgroup with c-KIT, NRAS, FLT3, CSF3R,

and TET2 mutations, WBC count and CR rate were not compared

between inv(16)/t(16;16) AML and t(8;21) AML patients in

previous studies. Our results showed that patients with

inv(16)/t(16;16) had significantly higher WBC counts than

those with t(8;21) in the context of c-KIT or NRAS mutations,

but no significant differences were found for CR rates.

Conclusion

This study has comprehensively analyzed the genetic mutations

of 134 CBF-AML patients to characterize certain crucial genetic

characteristics, compare the mutational profiles of t(8;21)

AML and inv(16)/t(16;16) AML, and establish unique genetic

maps. The major limitations of our study are the absence of

survival data, because these patients received treatment

in different medical institutions, and the fact that the

prognosis of these patients was affected by diverse factors

including physical status, financial situation, and the

different consolidation regimens that were administered.

The molecular mechanisms, exact characteristics, and clinical

implications of these mutations require further study.

Acknowledgments

This work was partly supported by grants from the Young

Scientists Foundation of Changzhou No. 2 People’s Hospital (No.

2019K002), the Science and Technology Project of the Changzhou

Health Committee (No. QN202035), the Science and Technology

Development Fund Project of Nanjing Medical University

(No. NMUB201), the Major Science Project of the Changzhou

Health Commission (No. ZD202018), the Project of Science and

Technology Support for Social Development of the Science and

Technology Bureau of Changzhou (No. CE20205027), and the

Changzhou Sci-Tech Program (No. CJ20210068).

Ethics

Ethics Committee Approval: The study was approved by the

local ethics committee and conducted in accordance with the

Declaration of Helsinki.

91


Qin W. et al: Mutation Profile in CBF-AML

Turk J Hematol 2022;39:84-93

Authorship Contributions

Surgical and Medical Practices: H.H.; Concept: N.J.; Design: W.Q.;

Data Collection or Processing: X.Ch.; Analysis or Interpretation:

H.J.S., , X.Ca.; Literature Search: Z.W.; Writing: N.J.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Schoch C, Kern W, Schnittger S, Büchner T, Hiddemann W, Haferlach T. The

influence of age on prognosis of de novo acute myeloid leukemia differs

according to cytogenetic subgroups. Haematologica 2004;89:1082-1090.

2. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le BMM,

Bloomfield CD, Cazzola M, Vardiman JW. The 2016 revision to the World

Health Organization classification of myeloid neoplasms and acute

leukemia. Blood 2016;127:2391-2405.

3. Döhner H, Estey E, Grimwade D, Amadori S,

Appelbaum FR, Büchner T, Dombret H, Ebert BL, Fenaux P, Larson RA, Levine

RL, Lo-Coco F, Naoe T, Niederwieser D, Ossenkoppele GJ, Sanz M, Sierra

J, Tallman MS, Tien HF, Wei AH, Löwenberg B, Bloomfield CD. Diagnosis

and management of AML in adults: 2017 ELN recommendations from an

international expert panel. Blood 2017;129:424-447.

4. Borthakur G, Kantarjian H. Core binding factor acute myelogenous

leukemia 2021 treatment algorithm. Blood Cancer J 2021;11:114.

5. Solh M, Yohe S, Weisdorf D, Ustun C. Core-binding factor acute

myeloid leukemia: heterogeneity, monitoring, and therapy. Am J

Hematol 2014;89:1121-1131.

6. Jourdan E, Boissel N, Chevret S, Delabesse E, Renneville A, Cornillet

P, Blanchet O, Cayuela JM, Recher C, Raffoux E, Delaunay J, Pigneux

A, Bulabois CE, Berthon C, Pautas C, Vey N, Lioure B, Thomas X, Luquet

I, Terré C, Guardiola P, Béné MC, Preudhomme C, Ifrah N, Dombret H;

French AML Intergroup. Prospective evaluation of gene mutations and

minimal residual disease in patients with core binding factor acute myeloid

leukemia. Blood 2013;121:2213-2223.

7. Schlenk RF, Benner A, Krauter J, Büchner T, Sauerland C, Ehninger G, Schaich

M, Mohr B, Niederwieser D, Krahl R, Pasold R, Döhner K, Ganser A, Döhner H,

Heil G. Individual patient data-based meta-analysis of patients aged 16 to

60 years with core binding factor acute myeloid leukemia: a survey of the

German Acute Myeloid Leukemia Intergroup. J Clin Oncol 2004;22:3741-

3750.

8. Marcucci G, Mrózek K, Ruppert AS, Maharry K, Kolitz JE, Moore JO, Mayer RJ,

Pettenati MJ, Powell BL, Edwards CG, Sterling LJ, Vardiman JW, Schiffer CA,

Carroll AJ, Larson RA, Bloomfield CD. Prognostic factors and outcome of core

binding factor acute myeloid leukemia patients with t(8;21) differ from

those of patients with inv(16): a Cancer and Leukemia Group B study. J Clin

Oncol 2005;23:5705-5717.

9. Downing JR. The core-binding factor leukemias: lessons learned from

murine models. Curr Opin Genet Dev 2003;13:48-54.

10. Quan X, Deng J. Core binding factor acute myeloid leukemia: advances

in the heterogeneity of KIT, FLT3, and RAS mutations (review). Mol Clin

Oncol 2020;13:95-100.

11. Duployez N, Marceau-Renaut A, Boissel N, Petit A, Bucci M, Geffroy S,

Lapillonne H, Renneville A, Ragu C, Figeac M, Celli-Lebras K, Lacombe C,

Micol JB, Abdel-Wahab O, Cornillet P, Ifrah N, Dombret H, Leverger G,

Jourdan E, Preudhomme C. Comprehensive mutational profiling of core

binding factor acute myeloid leukemia. Blood 2016;127:2451-2459.

12. Ishikawa Y, Kawashima N, Atsuta Y, Sugiura I, Sawa M, Dobashi N, Yokoyama

H, Doki N, Tomita A, Kiguchi T, Koh S, Kanamori H, Iriyama N, Kohno A,

Moriuchi Y, Asada N, Hirano D, Togitani K, Sakura T, Hagihara M, Tomikawa

T, Yokoyama Y, Asou N, Ohtake S, Matsumura I, Miyazaki Y, Naoe T, Kiyoi

H. Prospective evaluation of prognostic impact of KIT mutations on acute

myeloid leukemia with RUNX1-RUNX1T1 and CBFB-MYH11. Blood Adv

2020;4:66-75.

13. Lin LI, Chen CY, Lin DT, Tsay W, Tang JL, Yeh YC, Shen HL, Su FH,

Yao M, Huang SY, Tien HF. Characterization of CEBPA mutations

in acute myeloid leukemia: most patients with CEBPA mutations have

biallelic mutations and show a distinct immunophenotype of the leukemic

cells. Clin Cancer Res 2005;11:1372-1379.

14. Chen W, Konoplev S, Medeiros LJ, Koeppen H, Leventaki V, Vadhan-

Raj S, Jones D, Kantarjian HM, Falini B, Bueso-Ramos CE. Cuplike nuclei

(prominent nuclear invaginations) in acute myeloid leukemia are highly

associated with FLT3 internal tandem duplication and NPM1 mutation.

Cancer 2009;115:5481-5489.

15. Wang K, Zhou F, Cai X, Chao H, Zhang R, Chen S. Mutational landscape of

patients with acute myeloid leukemia or myelodysplastic syndromes in the

context of RUNX1 mutation. Hematology 2020;25:211-218.

16. Iriyama N, Hatta Y, Takeuchi J, Ogawa Y, Ohtake S, Sakura T, Mitani K, Ishida

F, Takahashi M, Maeda T, Izumi T, Sakamaki H, Miyawaki S, Honda S,

Miyazaki Y, Taki T, Taniwaki M, Naoe T. CD56 expression is an independent

prognostic factor for relapse in acute myeloid leukemia with t(8;21). Leuk

Res 2013;37:1021-1026.

17. Beaupre DM, Kurzrock R. RAS and leukemia: from basic mechanisms to

gene-directed therapy. J Clin Oncol 1999;17:1071-1079.

18. Simanshu DK, Nissley DV, McCormick F. RAS proteins and their regulators in

human disease. Cell 2017;170:17-33.

19. Faber ZJ, Chen X, Gedman AL, Boggs K, Cheng J, Ma J, Radtke I, Chao JR,

Walsh MP, Song G, Andersson AK, Dang J, Dong L, Liu Y, Huether R, Cai Z,

Mulder H, Wu G, Edmonson M, Rusch M, Qu C, Li Y, Vadodaria B, Wang J,

Hedlund E, Cao X, Yergeau D, Nakitandwe J, Pounds SB, Shurtleff S, Fulton

RS, Fulton LL, Easton J, Parganas E, Pui CH, Rubnitz JE, Ding L, Mardis ER,

Wilson RK, Gruber TA, Mullighan CG, Schlenk RF, Paschka P, Döhner K,

Döhner H, Bullinger L, Zhang J, Klco JM, Downing JR. The genomic landscape

of core-binding factor acute myeloid leukemias. Nat Genet 2016;48:1551-

1556.

20. Sood R, Hansen NF, Donovan FX, Carrington B, Bucci D,

Maskeri B, Young A, Trivedi NS, Kohlschmidt J, Stone RM,

Caligiuri MA, Chandrasekharappa SC, Marcucci G, Mullikin JC, Bloomfield CD,

Liu P. Somatic mutational landscape of AML with inv(16) or t(8;21) identifies

patterns of clonal evolution in relapse leukemia. Leukemia 2016;30:501-

504.

21. Boissel N, Leroy H, Brethon B, Philippe N, Botton S, Auvrignon A, Raffoux

E, Leblanc T, Thomas X, Hermine O, Quesnel B, Baruchel A, Leverger G,

Dombret H, Preudhomme C. Incidence and prognostic impact of c-Kit, FLT3,

and Ras gene mutations in core binding factor acute myeloid leukemia

(CBF-AML). Leukemia 2006;20:965-970.

22. Bowen DT, Frew ME, Hills R, Gale RE, Wheatley K, Groves MJ, Langabeer

SE, Kottaridis PD, Moorman AV, Burnett AK, Linch DC. RAS mutation in

acute myeloid leukemia is associated with distinct cytogenetic subgroups

but does not influence outcome in patients younger than 60 years. Blood

2005;106:2113-2119.

23. Yang X, Qian J, Sun A, Lin Jiang, Xiao G, Yin Jia, Chen S, Wu D. RAS mutation

analysis in a large cohort of Chinese patients with acute myeloid leukemia.

Clin Biochem 2013;46:579-583.

24. Bacher U, Haferlach T, Schoch C, Kern W, Schnittger S. Implications

of NRAS mutations in AML: a study of 2502 patients. Blood 2006;107:3847-

3853.

25. Kon A, Shih LY, Minamino M, Sanada M, Shiraishi Y, Nagata Y, Yoshida

K, Okuno Y, Bando M, Nakato R, Ishikawa S, Sato-Otsubo A, Nagae G,

Nishimoto A, Haferlach C, Nowak D, Sato Y, Alpermann T, Nagasaki M,

92


Turk J Hematol 2022;39:84-93

Qin W. et al: Mutation Profile in CBF-AML

Shimamura T, Tanaka H, Chiba K, Yamamoto R, Yamaguchi T, Otsu M,

Obara N, Sakata-Yanagimoto M, Nakamaki T, Ishiyama K, Nolte F, Hofmann

WK, Miyawaki S, Chiba S, Mori H, Nakauchi H, Koeffler HP, Aburatani

H, Haferlach T, Shirahige K, Miyano S, Ogawa S. Recurrent mutations in

multiple components of the cohesin complex in myeloid neoplasms. Nat

Genet 2013;45:1232-1237.

26. Thol F, Bollin R, Gehlhaar M, Walter C, Dugas M, Suchanek KJ, Kirchner A,

Huang L, Chaturvedi A, Wichmann M, Wiehlmann L, Shahswar R, Damm

F, Göhring G, Schlegelberger B, Schlenk R, Döhner K, Döhner H, Krauter J,

Ganser A, Heuser M. Mutations in the cohesin complex in acute myeloid

leukemia: clinical and prognostic implications. Blood 2014;123:914-920.

27. Sood R, Hansen NF, Donovan FX, Carrington B, Bucci D, Maskeri

B, Young A, Trivedi NS, Kohlschmidt J, Stone RM, Caligiuri MA,

Chandrasekharappa SC, Marcucci G, Mullikin JC, Bloomfield CD, Liu P.

Somatic mutational landscape of AML with inv(16) or t(8;21) identifies

patterns of clonal evolution in relapse leukemia. Leukemia 2016;30:501-

504.

28. Thota S, Viny AD, Makishima H, Spitzer B, Radivoyevitch T, Przychodzen B,

Sekeres MA, Levine RL, Maciejewski JP. Genetic alterations of the cohesin

complex genes in myeloid malignancies. Blood 2014;124:1790-1798.

29. Mazumdar C, Shen Y, Xavy S, Zhao F, Reinisch A, Li R, Corces MR, Flynn RA,

Buenrostro JD, Chan SM, Thomas D, Koenig JL, Hong WJ, Chang HY, Majeti

R. Leukemia associated cohesin mutants dominantly enforce stem cell

programs and impair human hematopoietic progenitor differentiation. Cell

Stem Cell 2015;17:675-688.

30. Park SH, Lee HJ, Kim IS, Kang JE, Lee EY, Kim HJ, Kim YK, Won JH, Bang

SM, Kim H, Song MK, Chung JS, Shin HJ. Incidences and prognostic impact

of c-KIT, WT1, CEBPA, and CBL mutations, and mutations associated with

epigenetic modification in core binding factor acute myeloid leukemia: a

multicenter study in a Korean population. Ann Lab Med 2015;35:288-297.

31. Jahn N, Terzer T, Sträng E, Dolnik A, Cocciardi S, Panina E, Corbacioglu A,

Herzig J, Weber D, Schrade A, Götze K, Schröder T, Lübbert M, Wellnitz D,

Koller E, Schlenk RF, Gaidzik VI, Paschka P, Rücker FG, Heuser M, Thol F,

Ganser A, Benner A, Döhner H, Bullinger L, Döhner K. Genomic heterogeneity

in core-binding factor acute myeloid leukemia and its clinical implication.

Blood Adv 2020;4:6342-6352.

93


RESEARCH ARTICLE

DOI: 10.4274/tjh.galenos.2021.2021.0203

Turk J Hematol 2022;39:94-102

Invasive Fungal Infections in Children with Leukemia: Clinical

Features and Prognosis

Lösemili Çocuklarda İnvazif Mantar Enfeksiyonları: Klinik Özellikler ve Prognoz

Melike Sezgin Evim 1 , Özlem Tüfekçi 2 , Birol Baytan 1 , Hale Ören 2 , Solmaz Çelebi 3 , Beyza Ener 4 , Kevser Üstün Elmas 5 ,

Şebnem Yılmaz 2 , Melek Erdem 2 , Mustafa Kemal Hacımustafaoğlu 3 , Adalet Meral Güneş 1

1Uludağ University Faculty of Medicine, Department of Pediatric Hematology, Bursa, Turkey

2Dokuz Eylül University Faculty of Medicine, Department of Pediatric Hematology, İzmir, Turkey

3Uludağ University Faculty of Medicine, Department of Pediatric Infection Disease, Bursa, Turkey

4Uludağ University Faculty of Medicine, Department of Medical Microbiology, Bursa, Turkey

5Uludağ University Faculty of Medicine, Department of Pediatrics, Bursa, Turkey

Abstract

Objective: The incidence of invasive fungal infections (IFIs) has

increased due to intensive chemotherapy in childhood leukemia. The

aim of this study was to evaluate the incidence, risk factors, causative

pathogens, and impact on survival of IFIs among pediatric leukemia

patients.

Materials and Methods: The hospital records of 307 children with

acute lymphoblastic leukemia (ALL, n=238), acute myeloid leukemia

(AML, n=51), and relapsed leukemia (n=18) between January 2010 and

December 2015 were retrospectively evaluated.

Results: A total of 1213 febrile neutropenia episodes were recorded

and 127 (10.4%) of them were related to an IFI. Of 307 children, 121

(39.4%) developed IFIs. The mean age was significantly older in the IFI

group compared to children without IFIs (p<0.001). IFIs were defined

as possible, probable, and proven in 73.2%, 11.9%, and 14.9% of

the attacks, respectively. Invasive aspergillosis (81.9%) was the most

frequent infection, followed by invasive candidiasis (13.4%) and rare

fungal diseases (4.8%). The majority of IFI attacks in both ALL and

AML occurred during the induction phase. In total, the death rate was

24% and the IFI-related mortality rate was 18%. The mortality rate

among children with IFIs was found to be significantly higher than

that of children without IFIs (p<0.001). Overall and event-free survival

rates at 5 years were also found to be significantly lower in the IFI

group (p<0.001). Relapse (odds ratio: 8.49) was the most effective risk

factor for mortality, followed by developing an IFI episode (odds ratio:

3.2) and AML (odds ratio: 2.33) according to multivariate regression

analysis.

Conclusion: Our data showed that IFIs were more common in older

children. Although proven and probable IFI episodes were more

frequently diagnosed in cases of relapse and AML, children with ALL

and AML had similar frequencies of experiencing at least one episode

Öz

Amaç: Çocukluk çağı lösemisinde yoğun kemoterapi nedeniyle invaziv

mantar enfeksiyonlarının (IFI) insidansı artmıştır. Bu çalışmanın amacı,

pediatrik lösemi hastalarında IFI’nın insidansını, risk faktörlerini,

nedensel patojenleri ve sağkalım üzerindeki etkisini değerlendirmektir.

Gereç ve Yöntemler: Ocak 2010 ile Aralık 2015 tarihleri arasında akut

lenfoblastik lösemili (ALL, n=238), akut myeloid lösemili (AML, n=51)

ve relaps lösemili (n=18) 307 çocukların hastane kayıtları geriye dönük

olarak değerlendirildi.

Bulgular: Toplam 1213 febril nötropeni atağı kaydedildi ve bunların

127’si (%10,4) IFI ile ilgiliydi. Bu 307 çocuğun 121’i (%39,4) IFI

geliştirdi. Ortalama yaş, IFI grubunda IFI olmayan çocuklara kıyasla

anlamlı olarak daha büyük bulundu (p<0,001). IFI, atakları sırasıyla;

%73,2, %11,9 ve %14,9’unda olası, yüksek olası ve kanıtlanmış olarak

tanımlandı. İnvazif aspergilloz (%81,9) en sık görülen enfeksiyondu,

bunu invaziv kandidiyazis (%13,4) ve nadir mantar hastalıkları (%4,8)

izledi. Hem ALL hem de AML’deki IFI ataklarının çoğu indüksiyon

aşamasında görüldü. Toplamda ölüm oranı %24 ve IFI bağlantılı ölüm

oranı %18 olarak bulundu. IFI’lı çocuklarda ölüm oranı, IFI olmayan

çocuklarda anlamlı olarak daha yüksek bulundu (p<0,001). Beş yılda

genel ve olaysız sağkalım, IFI grubunda önemli ölçüde daha düşük

bulundu (p<0,001). Relaps (odds oranı, 8,49), mortalite üzerinde en

etkili risk faktörü oldu ve bunu, çok değişkenli regresyon analizi ile

bir IFI epizodu geçirmek (odds oranı, 3,2) ve AML olmak (odds oranı,

2,33) izledi.

Sonuç: Çalışmamız, IFI’nın büyük yaştaki çocuklarda daha yaygın

olduğunu gösterdi. Kanıtlanmış ve olası IFI epizodlarının nüks ve

AML’de daha sık saptanmasına karşın, ALL ve AML’li çocukların da

benzer sıklıkta en az bir IFI atağı geçirdiği görüldü. Nadir mantar

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Melike Sezgin Evim, M.D., Uludağ University Faculty of Medicine,

Department of Pediatric Hematology, Bursa, Turkey

E-mail : melikevim@yahoo.com ORCID: orcid.org/0000-0002-4792-269X

Received/Geliş tarihi: March 25, 2021

Accepted/Kabul tarihi: November 17, 2021

94


Turk J Hematol 2022;39:94-102

Sezgin Evim M. et al: Fungal Infections in Leukemic Children

Abstract

of IFI. Rare fungal diseases were also identified as a major problem.

Despite success in treatment, IFIs increased the rate of mortality in

children with acute leukemia.

Keywords: Fungal infections, Pediatric leukemia, Acute lymphoblastic

leukemia, Acute myeloid leukemia, Febrile neutropenia

Öz

hastalıkları önemli bir sorun olarak tanımlandı. Tedavideki başarıya

rağmen, IFI’nın akut lösemili çocuklarda ölüm oranını artırdığı

saptandı.

Anahtar Sözcükler: Mantar enfeksiyonları, Pediatrik lösemi, Akut

lenfoblastik lösemi, Akut myeloid lösemi, Febril nötropeni

Introduction

The overall survival (OS) rate of pediatric acute leukemia has

improved over the years, reaching 90% for acute lymphoblastic

leukemia (ALL) and 70% for acute myeloid leukemia (AML) [1,2].

However, invasive fungal infections (IFIs) are still a significant

cause of mortality and morbidity in children with hematologic

malignancies [3]. It is hard to estimate the true incidence of

IFIs, but it has been increasing in recent years due to advances

in diagnostic methods, the use of intensive chemotherapy,

prolonged neutropenia, and the increased use of central

catheters [3,4]. Although new antifungals are available, IFIrelated

mortality still remains high, reported to be between 20%

and 70% in various studies [4,5,6,7,8].

The aim of this study was to evaluate the incidence, risk factors,

causative pathogens, and impact of IFIs on survival among

pediatric acute leukemia patients treated with Berlin-Frankfurt-

Munich (BFM) protocols in two major pediatric hematology

centers of Turkey.

Materials and Methods

The medical records of acute leukemia patients diagnosed

between 2010 and 2015 in the pediatric hematology

departments of Uludağ and Dokuz Eylül University Hospitals

were retrospectively evaluated. The study included only children

with ALL and AML, excluding hematopoietic stem cell recipients.

De nova ALL and AML patients were treated with BFM-based

protocols (ALL IC-BFM 2009 and AML-BFM 2004 and 2012).

ALL-REZ BFM 2002 was used for relapsed ALL and cases of

relapsed AML were treated according to AML-REZ BFM 2001/01.

A total of 307 patients’ data were eligible for evaluation. Febrile

neutropenia attacks were analyzed for defining and classifying

IFIs according to the European Organization for the Research

and Treatment of Cancer/Mycoses Study Group (EORTC/MSG)

classification system [9]. Based on this classification, probable

IFI requires the presence of a host factor, a clinical criterion,

and a mycological criterion. Cases that meet the criteria for

a host factor and the clinical criterion without a mycological

criterion are considered as possible IFI. Proven IFI is defined as

fungus detected by either histological analysis or culturing of a

specimen of tissue taken from a site of disease.

The patients were hospitalized in single rooms without highefficiency

particulate air filtered systems. Data including age,

gender, leukemia type, risk groups, treatment phase, duration of

neutropenia, and steroid use prior to the diagnosis of IFI were

collected from patients’ files. Neutropenia was defined as an

absolute neutrophil count (ANC) below 500/mm 3 and severe

neutropenia was defined as an ANC below 100/mm 3 . Bacterial

and fungal surveillances were performed for peripheral blood,

central venous catheters, throat, urine, and stool. Tissue biopsy

from the affected site for mycological examination was

evaluated when available. Chest high-resolution computed

tomography (HRCT) and abdominal ultrasound were performed

within the first week of persistent fever in spite of proper

antimicrobial treatment. In the presence of a localized finding

or persistent, prolonged fever, sinonasal and cranial imaging

(magnetic resonance imaging or computed tomography scan)

were also performed. Serum galactomannan (GM) levels were

measured twice weekly during febrile episodes using the Platelia

Aspergillus Enzyme Immunoassay Test (Bio-Rad). Samples with

an index greater than 0.5 in two consecutive measurements

were considered positive. The use of antifungal drugs for either

prophylaxis or treatment was recorded. The survival rates and

the risk factors affecting mortality in children with IFIs and

without IFIs were compared. The study was approved by the

relevant ethics committee on October 4, 2016 (decision number:

2016-17/12).

Statistical Analysis

All statistical analyses were carried out using IBM SPSS Statistics

23.0 (IBM Corp.). Descriptive data were presented as median and

minimum-maximum. Variables with non-normal distribution

were analyzed using the Mann-Whitney U and chi-square tests.

Risk factors for mortality were assessed using univariate and

multivariate logistic regression analysis. Values of p<0.05 were

considered statistically significant. Analyses for event-free

survival (EFS) and OS rates were performed according to the

Kaplan-Meier method, and survival curves were compared with

the log-rank test.

Results

The data of a total of 307 pediatric patients with acute leukemia

were retrospectively screened. Of these, 289 cases were de novo

95


Sezgin Evim M. et al: Fungal Infections in Leukemic Children

Turk J Hematol 2022;39:94-102

acute leukemia (ALL=238, AML=51) and the rest (n=18) were

relapsed cases. In total, 1213 febrile neutropenia episodes were

recorded and 127 (10.4%) of them were related to IFIs according

to the EORTC/MSG criteria. IFI developed in 121 children

(39.4%). The median age of the whole cohort was 78 (1.7-215.7)

months and it was found to be significantly higher in the IFI

group (n=121) compared to the children without IFIs (n=186)

(96 months vs. 58 months; p<0.001). The male-to-female ratio

was 1.47 (n=183/124). No significant difference was determined

in terms of gender (p>0.05).

Classification of IFIs

The distributions of patients and episodes of IFI are shown in

Table 1. According to the EORTC/MSG criteria, the episodes

were classified as proven (14.9%; n=19/127), probable (11.9%;

n=15/127), or possible (73.2%; n=93/127). In total, 35.2%

(n=84/238) of ALL, 39% (n=20/51) of AML, and 94.4% (n=17/18)

of relapsed patients experienced at least one episode of IFI. The

incidences of proven and probable IFI episodes were evaluated

together and found to be significantly higher in the relapsed

leukemia group compared to the de novo ALL and AML patients

(p<0.001).

Neutropenia was present in 82% (n=104/127) and severe

neutropenia was demonstrated in 73% (n=76/104) of all IFI

episodes. The median duration of neutropenia was 20 (1-78)

days. The group with proven or probable IFI together had a

significantly longer median duration of neutropenia than the

possible IFI cases (30 days vs. 16 days; p=0.002). The frequency

of IFI attacks in the ALL group was higher during induction

therapy (n=55/87), followed by high-risk blocks (n=18/87) and

the consolidation phase (n=14/87). In AML cases, IFIs usually

occurred during induction (72.7%, n=16/22).

Mycologic agents isolated during the episodes are given in

Table 2. The majority of IFI episodes were related to invasive

aspergillosis (IA) (81.9%), followed by invasive candidiasis (IC)

(13.4%) and rare fungal infections (4.8%). The features of rare

fungal infections are given in Table 3.

Generally, children with ALL in the high-risk group received

fluconazole prophylaxis while AML and relapse patients had

posaconazole or itraconazole. Access to posaconazole is

restricted over 13 years of age in Turkey. For younger children,

it could only be administered with the special approval of the

Turkish Ministry of Health. In the IFI group, 40% of the episodes

occurred under antifungal prophylaxis; the rates of IFI attacks

in ALL, AML, and relapsed leukemia cases were 30% (n=26/87),

50% (n=11/22), and 77% (n=14/18), respectively. Children with

proven or probable episodes were under antifungal prophylaxis

at the time of 21% (n=4/19) and 80% (n=12/15) of the episodes,

respectively. The most commonly used antifungal agents for

prophylaxis were fluconazole (42.9%), itraconazole suspension

(33.3%), and posaconazole (17.2%). Secondary antifungal

prophylaxis was administered for 63% (n=80/127) of the IFI

episodes and the major preferred agent was voriconazole (46%).

For the treatment of IFI episodes, voriconazole (38%, n=49/127),

caspofungin (22.8%, n=29/127), or liposomal amphotericin B

(11%, n=14/127) was given as a single agent. Combination

therapy was administered in 27% (n=35/127) of the attacks

when the disease was not taken under control by a single

agent. Caspofungin and voriconazole (n=13/35) were the most

commonly preferred combination therapy.

Outcome

In the whole cohort, the 5-year OS and EFS rates were found to

be 83.6% and 79% for ALL, 68.7% and 60.8% for AML, and 22%

and 17.6% for relapsed leukemia cases. In total, the mortality

rate was 24% (n=72/307). It was found to be significantly

Table 1. Distribution of patients and episodes of invasive fungal infections according to diagnosis.

Total, n ALL, % (n) AML, % (n)

Relapsed

leukemia, % (n)

Total patients 307 77.6% (238/307) 16.6% (51/307) 5.8% (18/307)

Patients without IFI 60.6% (186/307) 82.7% (154/186) 16.6% (31/186) 0.7% (1/186)

Patients with IFI 39.4% (121/307) 69.4% (84/121) 16.5% (20/121) 14.1% (17/121)

Total IFI episodes 127 68.5% (87/127) 17.3% (22/127) 14.2% (18/127)

Possible 73.2% (93/127) 80.5% (70/87) 68.2% (15/22) 44.5% (8/18)

Probable 11.9% (15/127) 8% (7/87) 9.1% (2/22) 33.3% (6/18)

Proven 14.9% (19/127) 11.5% (10/87) 22.7% (5/22) 22.2% (4/18)

Proven + probable 28.8% (34/127) 19.5% (17/87) 31.8% (7/22) 55.5% (10/18)*

ALL: Acute lymphoblastic leukemia, AML: acute myeloid leukemia, IFI: invasive fungal infection, *: p<0.001.

96


Turk J Hematol 2022;39:94-102

Sezgin Evim M. et al: Fungal Infections in Leukemic Children

higher in the IFI group (34%, n=41/121) compared to the non-

IFI group (16.6%, n=31/186) (p<0.001). IFI-related mortality was

observed in 18% (n=22/121) of cases (Figure 1). The mortality

rate in children with IFIs increased to 52.9% (n=18/34) when

possible IFI attacks were excluded. Five-year EFS and OS rates

were found to be significantly lower in the IFI group than in

cases without IFIs (EFS: 79.6% vs. 62.7%, OS: 83.9% vs. 67.5%)

(p<0.001) (Figure 2). Both EFS and OFS at 5 years in the IFI group

were found to be even lower when possible IFI attacks were

removed from the analysis (47.1%).

Table 2. Classification according to the diagnosis of invasive fungal infection.

Diagnosis of IFI

Episodes of IFI

(n=127)

Proven IFI,

14.9% (n=19)

Probable IFI,

11.9% (n=15)

Possible IFI,

73.2% (n=93)

Invasive aspergillosis 81.9% (n=104) 3 15 86

IPA 76.3% (n=97) - 15 82

SNIA 4.8% (n=6) 3* - 3

CNSIA 0.8% (n=1) - - 1

Invasive candidiasis 13.4% (n=17) 10** - 7

HSC 7.8% (n=10) 3 - 7

Candidemia 6.3% (n=7) 7 - -

Rare fungal inf. 4.8% (n=6)*** 6 - -

Mucorales (n=3) 3 - -

Fusarium (n=1) 1 - -

Dematiaceous (n=1) 1 - -

Alternaria (n=1) 1 - -

IFI: Invasive fungal infection, IPA: invasive pulmonary aspergillosis, SNIA: sinonasal invasive aspergillosis, CNSIA: central nervous system invasive aspergillosis, HSC: hepatosplenic

candidiasis.

*: Biopsy proven from sinonasal cavity: A. flavus (n=3).

**: Isolated from blood cultures: C. parapsilosis (n=6), C. albicans (n=3), C. guilliermondii (n=1).

***: Biopsy proven from sinonasal cavity (n=5), left maxillectomy (n=1).

Table 3. Features of rare fungal infections.

Rare fungal

infection

Diagnosis

Age,

months

Primary

prophylaxis

Biopsy region Treatment Status

Mucorales Relapsed AML 40 Posaconazole Left maxillectomy

Mucorales ALL-HRG 204 No Sinonasal cavity

Mucorales ALL-HRG 40 No Sinonasal cavity

Fusarium Relapsed ALL 196 Posaconazole Sinonasal cavity

Liposomal

amphotericin B

Liposomal

amphotericin B

Liposomal

amphotericin B

Liposomal

amphotericin B

Death due to

resistant disease

Death due to IFI

Death due to IFI

Death due to IFI

Dematiaceous AML 52 No Sinonasal cavity Caspofungin Alive

Alternaria ALL 182 No Sinonasal cavity

ALL: Acute lymphoblastic leukemia, AML: acute myeloid leukemia, HRG: high-risk group, IFI: invasive fungal infection.

Liposomal

amphotericin B

Alive

97


Sezgin Evim M. et al: Fungal Infections in Leukemic Children

Turk J Hematol 2022;39:94-102

Factors Affecting Mortality

For 307 children, the risk factors affecting mortality were

evaluated by univariate regression analysis. This revealed

that high-risk ALL (n=30) and AML (n=51), relapse (n=18),

neutropenia longer than 10 days (n=67), development of an

IFI episode, and presence of proven or probable IFI significantly

increased the rate of mortality (p<0.001). Gender and age at

diagnosis did not have any effect (p>0.05).

Independent risk factors for mortality were determined by

multivariate regression analysis (Table 4). Relapse (odds ratio:

8.49) was the most effective risk factor for mortality, followed

by development of an IFI episode (odds ratio: 3.2) and AML

(odds ratio: 2.33).

Discussion

We investigated a large sample of IFI data in children with

acute leukemia, which provided a valuable assessment of IFI

Figure 1. The rate and causes of mortality.

Figure 2. a). Event-free survival, b) Overall survival.

IFI: Invasive fungal infection

Table 4. Factors affecting mortality according to multivariate regression analysis.

Β p Odds ratio 95% CI

Relapse 2.14 <0.001 8.49 3.98-18.1

IFI episode 1.18 0.001 3.2 1.58-6.75

Leukemia type (AML) 0.84 0.02 2.33 1.1-4.92

AML: Acute myeloid leukemia, IFI: invasive fungal infection, CI: confidence interval.

98


Turk J Hematol 2022;39:94-102

Sezgin Evim M. et al: Fungal Infections in Leukemic Children

epidemiology and outcomes in our country. In the current

study, the incidence of IFI episodes was found to be 10.4%.

The incidence in cases of hematologic malignancies ranged

between 1.7% and 35.4% in various studies [4,9,10,11,12].

However, it may increase further in autopsy findings [13]. The

wide range among studies could be explained by differences

in study populations, hospital conditions, usage of prophylactic

antifungal agents, and the criteria used in defining IFI. The

current study included only children with acute leukemia, and

IFI attacks were defined according to EORTC/MSG criteria. The

majority of attacks were in the group with possible IFIs (73.2%),

since the use of invasive techniques such as bronchoalveolar

lavage (BAL) was limited. The classification was mainly based

on host factors, clinical criteria, imaging techniques, and serum

GM levels. However, some of the cases within the possible group

might be confirmed as Mucorales or other fungal diseases if

histopathologic diagnosis methods are available, similarly to a

case reported in the literature [14]. Among the present study

group, proven and probable attacks were determined in 14.9%

and 11.9% of the cases, respectively. Proven and probable IFI

incidences in recent studies with BFM groups were reported

to be lower than 10% [15,16]. Unlike our study, those two

studies included not only hematologic malignancies but

also hematopoietic stem cell transplantation recipients and

antifungal prophylaxis was administered to the majority of

them.

The mean age in the IFI group was significantly higher compared

to children in the non-IFI group. The incidence of IFIs is reported

to increase with age [4,7,17]. This finding could be attributed

to the fact that colonization is less frequent in the younger

population due to less contact with fungal spores and the lower

frequency of unfavorable genetic mutations [1,18].

We found that children with ALL and AML had a similar

frequency of experiencing at least one episode of IFI. This finding

is contrary to the previous data in the literature suggesting that

the frequency of IFI episodes in AML is higher than that in ALL

[4,19]. The prevalence of IFI in ALL is reported to be between

4% and 35% depending on the era, chemotherapy protocol, risk

categories, and antifungal prophylaxis [6,7,12,17,20,21,22]. In

the current study, only 30% of children with ALL were receiving

antifungal prophylaxis and the majority of them developed IFI

during induction therapy or high-risk blocks, which are typically

associated with severe neutropenia and high-dose steroids.

Prolonged and severe neutropenia was defined as a determinant

for IFI [3,4]. In our study, proven and probable IFI attacks also

had a significantly longer mean duration of neutropenia than

possible IFI attacks (p=0.002). Similarly, reports from different

countries showed that the majority of IFI attacks in both ALL

and AML cases occurred during the induction phase and with

more intensive chemotherapy protocols [12,20,23,24,25].

In developing countries, ALL patients receiving intensive

chemotherapy phases may benefit from antifungal prophylaxis

for decreasing IFI episodes in these patients.

The most common cause of IFI episodes in our study was IA

(81.9%), and the majority were cases of invasive pulmonary

aspergillosis (IPA) (76.3%). Aspergillus flavus was isolated

from the sinonasal cavity in 3 out of 8 biopsy materials. After

excluding possible IFI episodes, the proven and probable

incidence of IA was 17%. The incidences of IA and IPA have

been increasing in patients with pediatric leukemia since 2000

with the use of new diagnostic methods [4,26]. Crassard et al.

[5] showed that BAL results were suggestive of IPA in 92% of

children with pediatric leukemia. However, invasive procedures

like BAL and biopsy are generally avoided in children due to

thrombocytopenia and the risk of bleeding. Instead of biopsy,

the combination of chest HRCT and serum GM level is more

widely used for non-invasive diagnosis and it is also the

preferred approach in Turkey. However, GM levels may not be

elevated in localized IA, especially under antifungal prophylaxis

or empiric antifungal treatment [27,28].

The frequency of IC in total IFI episodes was found to be 13.4%.

However, it increased to 52.6% (n=10/19) among the proven IFI

episodes. In our series, the incidence of hepatosplenic candidiasis

(HSC) was determined as 7.8% and Candida spp. were isolated

from blood cultures in 30% of those cases. Celkan et al. [29],

in a multicentric study from Turkey, recently reported that 8

out of 40 (22.5%) children with HSC had Candida spp. and

only one case was C. albicans. In the current study, the most

commonly isolated pathogen was also non-albicans, namely C.

parapsilosis (Table 2). It appears that C. albicans, which used to

be the most commonly reported pathogen, has been replaced

by C. parapsilosis and other non-albicans species [6,12,19,30].

The current data showed a high rate of rare fungal infections,

which were detected in 6 of 127 IFI episodes (4.8%). However,

this incidence was increased to 31.5% (n=6/19) among proven

attacks. Epidemiological data for children regarding rare fungal

infections are limited [14,31,32,33,34]. Three out of 6 children

had mucormycosis and the others had fusarium, dematiaceous,

and alternaria involvements. The largest registry for invasive

mucormycosis in children showed that its frequency was

higher in cases of hematologic malignancies compared to

other malignancies and a variety of other disorders [31,32,33].

A retrospective study from Italy also reported that underlying

hematologic malignancies, particularly acute leukemia and

lymphoma, are risk factors for developing mucormycosis [34].

Two patients with mucormycosis and one with fusarium died

despite effective antifungal treatment combined with surgical

resection. The mortality rates for both mucormycosis and

fusarium are reported to be high [31,33]. Limited data related

to dematiaceous and alternaria cases in children with leukemia

are available in the literature [35,36]. In our study, these cases

99


Sezgin Evim M. et al: Fungal Infections in Leukemic Children

Turk J Hematol 2022;39:94-102

were successfully cured. The most important prognostic factor

is early clinical suspicion, timely aggressive systemic antifungal

treatment, and surgical procedures.

In the present study, the mortality rate for the whole cohort

was 24% and the rate of deaths attributable to IFI was found to

be 18%. IFI-related mortality in cases of hematologic disorders

ranged from 5% to 14% in Turkey [10,12,37]. It is also reported

to be as high as 21% to 48% in children with leukemia [7,21,38].

The current study also separately analyzed the mortality rates

for IA, IC, and rare fungal infections, which were found to be

16%, 17%, and 50%, respectively. The high mortality rate in the

rare fungal infection subgroup still remains an important issue

[19,31]. Although mortality rates due to IFIs were reported high

in the 1990s, they significantly decreased with the development

of early diagnostic tools and new antifungal agents [3,5,26,39].

Our data showed that OS and EFS were significantly lower in

children with IFIs than those without IFIs. Multivariate regression

analyses revealed that independent risk factors affecting

mortality were recurrent disease, development of an IFI episode,

and AML, which increased the mortality rate 8.4, 3.2, and 2.3

times, respectively. Data on the impact of IFIs on survival rates

in children with hematologic malignancies are rare. There are

three major studies reported regarding this issue. Two of them

did not find any differences in survival rates [40,41]. However,

those studies included small numbers of children. Similar to

our findings, Kobayashi et al. [7,42] found significantly lower

survival in patients with IFIs compared to those without IFI in a

patient population including hematologic malignancies, other

malignant diseases, and aplastic anemia.

Study Limitations

The data were retrospectively collected. The classification

was mainly based on host factors, clinical criteria, imaging

techniques, and GM levels. BAL was not administered for any of

the patients. Therefore, most of the attacks were in the “possible”

group. However, it may have been seen that these cases were

caused by other fungal pathogens if invasive techniques were

available.

Conclusion

Proven and probable IFI episodes concurrently occurred in ALL,

AML, and relapsed cases at rates of 19.5%, 31.8%, and 55.5%,

respectively. Although the majority of attacks were related to

IA, rare fungal infections were also isolated in almost 5% of

patients. Prolonged severe neutropenia was one of the major

risk factors for IFIs. Multivariate regression analysis showed

that IFIs significantly increased mortality, and the survival rates

were significantly lower in patients with IFIs. Our study clearly

shows that IFIs are poor prognostic factors in children with

hematologic malignancies.

Ethics

Ethics Committee Approval: The study was approved by the

relevant ethics committee on October 4, 2016 (decision number

2016-17/12).

Informed Consent: Obtained.

Authorship Contributions

Concept: M.S.E., H.Ö., A.M.G., B.B., Ş.Y.; Data collection or

processing: Ö.T., B.B., K.Ü.E., B.E., S.Ç., Ş.Y., M.E., M.K.H.; Analysis

or interpretation: M.S.E., Ö.T., H.Ö., A.M.G., B.E., K.Ü.E., S.Ç.,

M.K.H.; Literature review: M.S.E., H.Ö., K.Ü.E., A.M.G.; Writing:

M.S.E., H.Ö., A.M.G.

Conflicts of Interest: No conflicts of interest were declared by

the authors.

Financial Disclosure: The authors declared that this study

received no financial support.

Acknowledgments: The authors declared that they had no

financial, consulting, or personal relationships with other

people or organizations. This study was presented orally at the

44 th National Hematology Congress.

References

1. Pui CH, Yang JJ, Hunger SP, Pieters R, Schrappe M, Biondi A, Vora A, Baruchel

A, Silverman LB, Schmiegelow K, Escherich G, Horibe K, Benoit YC, Izraeli

S, Yeoh AE, Liang DC, Downing JR, Evans WE, Relling MV, Mullighan CG.

Childhood acute lymphoblastic leukemia: progress through collaboration. J

Clin Oncol 2015;33:2938-2948.

2. Hasle H, Kaspers GJ. Strategies for reducing the treatment-related physical

burden of childhood acute myeloid leukaemia - a review. Br J Haematol

2017;176:168-178.

3. Groll AH, Castagnola E, Cesaro S, Dalle JH, Engelhard D, Hope W, Roilides

E, Styczynski J, Warris A, Lehrnbecher T; Fourth European Conference on

Infections in Leukaemia; Infectious Diseases Working Party of the European

Group for Blood Marrow Transplantation (EBMT-IDWP); Infectious Diseases

Group of the European Organisation for Research and Treatment of Cancer

(EORTC-IDG); International Immunocompromised Host Society (ICHS);

European Leukaemia Net (ELN). Fourth European Conference on Infections

in Leukaemia (ECIL-4): guidelines for diagnosis, prevention, and treatment

of invasive fungal diseases in paediatric patients with cancer or allogeneic

haemopoietic stem-cell transplantation. Lancet Oncol 2014;15:e327-e340.

4. Ruijters VJ, Oosterom N, Wolfs TFW, van den Heuvel-Eibrink MM, van

Grotel MJ. Frequency and determinants of invasive fungal infections in

children with solid and hematologic malignancies in a nonallogeneic stem

cell transplantation setting: a narrative review. Pediatr Hematol Oncol

2019;41:345-354.

5. Crassard N, Hadden H, Piens MA, Pondarré C, Hadden R, Galambrun C,

Pracros JP, Souillet G, Basset T, Berthier JC, Philippe N, Bertrand Y. Invasive

aspergillosis in a paediatric haematology department: a 15-year review.

Mycoses 2008;51:109-116.

6. Mor M, Gilad G, Kornreich L, Fisher S, Yaniv I, Levy I. Invasive fungal

infections in pediatric oncology. Pediatr Blood Cancer 2011;56:1092-1097.

7. Kobayashi R, Kaneda M, Sato T, Ichikawa M, Suzuki D, Ariga T. The clinical

feature of invasive fungal infection in pediatric patients with hematologic

100


Turk J Hematol 2022;39:94-102

Sezgin Evim M. et al: Fungal Infections in Leukemic Children

and malignant diseases: a 10-year analysis at a single institution at Japan. J

Pediatr Hematol Oncol 2008;30:886-890.

8. Lashkari HP, Fernandes N, Alva K, Rai S. Central nervous system fungal

infection and acute lymphoblastic leukemia in children: what is the optimal

duration of antifungal therapy? J Pediatr Hematol Oncol 2017;39:e312-e317.

9. De Pauw B, Walsh TJ, Donnelly JP, Stevens DA, Edwards JE, Calandra T,

Pappas PG, Maertens J, Lortholary O, Kauffman CA, Denning DW, Patterson

TF, Maschmeyer G, Bille J, Dismukes WE, Herbrecht R, Hope WW, Kibbler

CC, Kullberg BJ, Marr KA, Muñoz P, Odds FC, Perfect JR, Restrepo A,

Ruhnke M, Segal BH, Sobel JD, Sorrell TC, Viscoli C, Wingard JR, Zaoutis T,

Bennett JE; European Organization for Research and Treatment of Cancer/

Invasive Fungal Infections Cooperative Group; National Institute of Allergy

and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus

Group. Revised definitions of invasive fungal disease from the European

Organization for Research and Treatment of Cancer/Invasive Fungal

Infections Cooperative Group and the National Institute of Allergy and

Infectious Diseases Mycoses Study Group EORTC/MSG) Consensus Group.

Clin Infect Dis 2008;46:1813-1821.

10. Kaya Z, Gursel T, Kocak U, Aral YZ, Kalkanci A, Albayrak M.

Invasive fungal infections in pediatric leukemia patients receiving

fluconazole prophylaxis. Pediatr Blood Cancer 2009;52:470-475.

11. Tüfekçi Ö, Yılmaz Bengoa Ş, Demir Yenigürbüz F, Şimşek E, Hilkay Karapınar

T, İrken G, Ören H. Management of invasive fungal infections in pediatric

acute leukemia and the appropriate time for restarting chemotherapy. Turk

J Hematol 2015;32:329-337.

12. Sahbudak Bal Z, Yilmaz Karapinar D, Karadas N, Sen S, Onder Sivis Z, Akinci

AB, Balkan C, Kavakli K, Vardar F, Aydinok Y. Proven and probable invasive

fungal infections in children with acute lymphoblastic leukaemia: results

from an university hospital, 2005-2013. Mycoses 2015;58:225-232.

13. Lehrnbecher T, Frank C, Engels K, Kriener S, Groll AH, Schwabe D. Trends in

the postmortem epidemiology of invasive fungal infections at a university

hospital. J Infect 2010;61:259-265.

14. Kebudi R, Kızılocak H, Hafız G, Erturan Z. Successful outcome of

mucormycosis in a child with acute lymphoblastic leukemia. Turk Pediatri

Ars 2020;55:207-209.

15. Lehrnbecher T, Schöning S, Poyer F, Georg J, Becker A, Gordon K, Attarbaschi

A, Groll AH. Incidence and outcome of invasive fungal diseases in children

with hematological malignancies and/or allogeneic hematopoietic stem cell

transplantation: results of a prospective multicenter study. Front Microbiol

2019;10:681.

16. Cesaro S, Tridello G, Castagnola E, Calore E, Carraro F, Mariotti I, Colombini

A, Perruccio K, Decembrino N, Russo G, Maximova N, Baretta V, Caselli D.

Retrospective study on the incidence and outcome of proven and probable

invasive fungal infections in high-risk pediatric onco-hematological

patients. Eur J Haematol 2017;99:240-248.

17. Rosen GP, Nielsen K, Glenn S, Abelson J, Deville J, Moore TB. Invasive fungal

infections in pediatric oncology patients: 11-year experience at a single

institution. J Pediatr Hematol Oncol 2005;27:135-140

18. Dini G, Castagnola E, Comoli P, van Tol MJ, Vossen JM. Infections after stem

cell transplantation in children: state of the art and recommendations.

Bone Marrow Transplant 2001;28(Suppl 1):S18-21.

19. Bartlett AW, Cann MP, Yeoh DK, Bernard A, Ryan AL, Blyth CC, Kotecha RS,

McMullan BJ, Moore AS, Haeusler GM, Clark JE. Epidemiology of invasive

fungal infections in immunocompromised children; an Australian national

10-year review. Pediatr Blood Cancer 2019;66:e27564.

20. Wang SS, Kotecha RS, Bernard A, Blyth CC, McMullan BJ, Cann MP, Yeoh

DK, Bartlett AW, Ryan AL, Moore AS, Bryant PA, Clark J, Haeusler GM.

Invasive fungal infections in children with acute lymphoblastic leukaemia:

results from four Australian centres, 2003-2013. Pediatr Blood Cancer

2019;66:e27915.

21. Castagnola E, Cesaro S, Giacchino M, Livadiotti S, Tucci F, Zanazzo G, Caselli

D, Caviglia I, Parodi S, Rondelli R, Cornelli PE, Mura R, Santoro N, Russo G,

De Santis R, Buffardi S, Viscoli C, Haupt R, Rossi MR. Fungal infections in

children with cancer: a prospective, multicenter surveillance study. Pediatr

Infect Dis J 2006;25:634-639.

22. Hale KA, Shaw PJ, Dalla-Pozza L, MacIntyre CR, Isaacs D, Sorrell TC.

Epidemiology of paediatric invasive fungal infections and a case-control

study of risk factors in acute leukaemia or post stem cell transplant. Br J

Haematol 2010;149:263-272.

23. Inaba H, Pei D, Wolf J, Howard SC, Hayden RT, Go M, Varechtchouk O, Hahn

T, Buaboonnam J, Metzger ML, Rubnitz JE, Ribeiro RC, Sandlund JT, Jeha S,

Cheng C, Evans WE, Relling MV, Pui CH. Infection-related complications

during treatment for childhood acute lymphoblastic leukemia. Ann Oncol

2017;28:386-392.

24. Sun Y, Huang H, Chen J, Li J, Ma J, Li J, Liang Y, Wang J, Li Y, Yu K, Hu J,

Jin J, Wang C, Wu D, Xiao Y, Huang X. Invasive fungal infection in patients

receiving chemotherapy for hematological malignancy: a multicenter,

prospective, observational study in China. Tumour Biol 2015;36:757-767.

25. Korula A, Abraham A, Abubacker FN, Viswabandya A, Lakshmi KM, Abraham

OC, Rupali P, Varghese GM, Michael JS, Srivastava A, Mathews V, George

B. Invasive fungal infection following chemotherapy for acute myeloid

leukaemia-Experience from a developing country. Mycoses 2017;60:686-

691.

26. Lass-Flörl C. The changing face of epidemiology of invasive fungal disease

in Europe. Mycoses 2009;52:197-205.

27. Kostamo K, Richardson M, Eerola E, Rantakokko-Jalava K, Meri T, Malmberg

H, Toskala E. Negative impact of Aspergillus galactomannan and DNA

detection in the diagnosis of fungal rhinosinusitis. J Med Microbiol

2007;56(Pt 10):1322-1327.

28. Maertens J, Theunissen K, Lodewyck T, Lagrou K, Van Eldere J. Advances in

the serological diagnosis of invasive Aspergillus infections in patients with

haematological disorders. Mycoses 2007;50(Suppl 1):2-17.

29. Celkan T, Kizilocak H, Evim M, Meral Güneş A, Özbek NY, Yarali N, Ünal E,

Patiroğlu T, Yilmaz Karapinar D, Sarper N, Zengin E, Karaman S, Koçak Ü,

Kürekçi E, Özdemir C, Tuğcu D, Uysalol E, Dikme G, Adaletli İ, Kuruoğlu

S, Kebudi R. Hepatosplenic fungal infections in children with leukemiarisk

factors and outcome: a multicentric study. J Pediatr Hematol Oncol

2019;41:256-260.

30. Zaoutis T. Candidemia in children. Curr Med Res Opin 2010;26:1761-1768.

31. Pana ZD, Seidel D, Skiada A, Groll AH, Petrikkos G, Cornely OA, Roilides

E; Collaborators of Zygomyco.net and/or FungiScope Registries. Invasive

mucormycosis in children: an epidemiologic study in European and non-

European countries based on two registries. BMC Infect Dis 2016;16:667.

32. Candoni A, Klimko N, Busca A, Di Blasi R, Shadrivova O, Cesaro S, Zannier

ME, Verga L, Forghieri F, Calore E, Nadali G, Simonetti E, Muggeo P, Quinto

AM, Castagnola C, Cellini M, Del Principe MI, Fracchiolla N, Melillo L,

Piedimonte M, Zama D, Farina F, Giusti D, Mosna F, Capelli D, Delia M,

Picardi M, Decembrino N, Perruccio K, Vallero S, Aversa F, Fanin R, Pagano L;

SEIFEM Group (Epidemiological Surveillance of Infections in Haematological

Diseases). Fungal infections of the central nervous system and paranasal

sinuses in onco-haematologic patients. Epidemiological study reporting

the diagnostic-therapeutic approach and outcome in 89 cases. Mycoses

2019;62:252-260.

33. Arnoni MV, Paula CR, Auler ME, Simões CCN, Nakano S, Szeszs MW, Melhem

MSC, Pereira VBR, Garces HG, Bagagli E, Silva EG, de Macêdo MF, Ruiz LDS.

Infections caused by Fusarium species in pediatric cancer patients and

review of published literature. Mycopathologia 2018;183:941-949.

34. Muggeo P, Calore E, Decembrino N, Frenos S, De Leonardis F, Colombini A,

Petruzziello F, Perruccio K, Berger M, Burnelli R, Zanazzo GA, Santoro N,

Cesaro S. Invasive mucormycosis in children with cancer: a retrospective

study from the Infection Working Group of Italian Pediatric Hematology

Oncology Association. Mycoses 2019;62:165-170.

101


Sezgin Evim M. et al: Fungal Infections in Leukemic Children

Turk J Hematol 2022;39:94-102

35. Astolfo MF, Cañazares P, Majek E, Burgesser V, Caruso M, Basco J, Alvarado

C, Carnovale S. Invasive acute sinusitis by Exserohilum rostratum in a

patient with medullary relapse of acute lymphoblastic leukemia. Arch

Argent Pediatr 2018;116:e594-e598.

36. Maloney AM, Ethier MC, Mitchell D, Zaoutis T, Sung L. Childhood Acute

Myeloid Leukemia Infection Research Group. Alternaria sinusitis

in children with acute myeloid leukemia: case reports from the Childhood

Acute Myeloid Leukemia Infection Research Group. Leuk Lymphoma

2010;51:345-347.

37. Baytan B, Güneş AM, Çelebi S, Günay Ü. Invasive fungal diseases in children

with hematologic disorders. Turk J Hematol 2009;26:190-196.

38. Blade J, Lopez-Guillermo A, Rozman C, Grañena A, Bruguera M, Bordas J,

Cervantes F, Carreras E, Sierra J, Montserrat E. Chronic systemic candidiasis

in acute leukemia. Ann Hematol 1992;64:240-244.

39. Groll AH, Kurz M, Schneider W, Witt V, Schmidt H, Schneider M, Schwabe

D. Five-year-survey of invasive aspergillosis in a paediatric cancer centre.

Epidemiology, management and long-term survival. Mycoses 1999;42:431-

442.

40. Ducassou S, Rivaud D, Auvrignon A, Vérité C, Bertrand Y, Gandemer V,

Leverger G. Invasive fungal infections in pediatric acute myelogenous

leukemia. Pediatr Infect Dis J 2015;34:1262-1264.

41. Kumar J, Singh A, Seth R, Xess I, Jana M, Kabra SK. Prevalence and

predictors of invasive fungal infections in children with persistent febrile

neutropenia treated for acute leukemia - a prospective study. Indian J

Pediatr 2018;85:1090-1095.

42. Kobayashi R, Hori D, Sano H, Suzuki D, Kishimoto K, Kobayashi K. Risk factors

for invasive fungal infection in children and adolescents with hematologic

and malignant diseases: a 10-year analysis in a single institute in Japan.

Pediatr Infect Dis J 2018;37:1282-1285.

102


RESEARCH ARTICLE

DOI: 10.4274/tjh.galenos.2022.2021.0675

Turk J Hematol 2022;39:103-108

Eltrombopag for Treatment of Thrombocytopenia Following

Hematopoietic Stem Cell Transplantation

Hematopoetik Kök Hücre Nakli Sonrası Trombositopeni Tedavisinde Eltrombopag Kullanımı

Zeynep Tuğba Güven 1 , Serhat Çelik 1 , Bülent Eser 2 , Mustafa Çetin 1 , Ali Ünal 1 , Leylagül Kaynar 1

1Erciyes University Faculty of Medicine, Department of Hematology, Kayseri, Turkey

2Medical Park Hospital, Antalya, Turkey

Abstract

Objective: This study aimed to evaluate the efficacy and safety of

eltrombopag (ELT) in the treatment of thrombocytopenia following

hematopoietic stem cell transplantation (HSCT).

Materials and Methods: Forty-eight patients treated with ELT for

thrombocytopenia after allogeneic or autologous transplantation at

the Erciyes University Bone Marrow Transplantation Center between

July 2017 and July 2021 were evaluated retrospectively.

Results: Forty-eight HSCT recipients were included in this study.

Thirty (62.5%) patients were evaluated as having experienced delayed

platelet recovery (DPR) and 18 (37.5%) patients as having experienced

secondary failure of platelet recovery (SFPR). The median platelet

count before ELT treatment was 13x10 9 /L (range: 3-20x10 9 /L). Twentythree

patients responded to treatment and the cumulative incidence

of successful platelet recovery was 48%. Patients with both DPR

and SFPR responded, but patients with DPR had a higher response

rate (50% vs. 44%). The median platelet count of the 23 responding

patients was 12x10 9 /L (5-19x10 9 /L) before treatment and 68x10 9 /L

(52-266x10 9 /L) after treatment (p<0.0001). While the number of bone

marrow megakaryocytes before treatment was adequate in 22 (46%)

cases, it was decreased in 26 (54%) cases. Patients with adequate bone

marrow megakaryocytes had a better response rate than those without

(77% vs. 23%, p<0.0001). The group with adequate megakaryocytes

responded to treatment at a median of 33 days (range: 9-174 days).

Patients with decreased megakaryocytes responded at a median of

55 days (30-164 days) (p=0.002). No drug-related side effects were

observed in any patients.

Conclusion: This real-life experience demonstrates that ELT is an

effective and safe treatment option for thrombocytopenia after HSCT.

The adequacy of bone marrow megakaryocytes before ELT treatment

was an important factor affecting response to treatment.

Keywords: Eltrombopag, Thrombocytopenia, Hematopoietic stem cell

transplantation, Platelet recovery

Öz

Amaç: Transplantasyon sonrası trombositopeni tedavisinde

eltrombopagın (ELT) etkinlik ve güvenliğini değerlendirmeyi amaçladık.

Gereç ve Yöntemler: Erciyes Üniversitesi Kemik İliği Nakil Merkezi’nde

Temmuz 2017-Temmuz 2021 tarihleri arasında allojenik veya otolog

transplantasyon sonrası trombositopeni nedeniyle ELT ile tedavi edilen

48 hasta retrospektif olarak değerlendirildi.

Bulgular: Bu çalışmaya 48 hematopoietik kök hücre nakli (HKHN)

alıcısı dahil edildi. Otuz (%62,5) hasta gecikmiş trombosit iyileşmesi

(DPR) ve 18 (%37,5) hasta ikincil trombosit iyileşme başarısızlığı (SFPR)

olarak değerlendirildi. ELT tedavisinden önce medyan trombosit sayısı

13x10 9 /L (aralık, 3-20x10 9 /L) idi. Yirmi üç hasta tedaviye yanıt verdi ve

başarılı trombosit iyileşmesinin kümülatif insidansı %48 idi. Hem DPR

hem de SFPR’li hastalar yanıt verdi, ancak DPR’li hastalarda yanıt oranı

daha yüksekti (%50’ye karşı %44). Yanıt veren 23 hastanın medyan

trombosit sayısı tedaviden önce ve sonra 12 (5-19)x10 9 /L ve 68 (52-

266)x10 9 /L idi (p<0,0001). Tedavi öncesi kemik iliği megakaryosit sayısı

22 (%46) hastada yeterli iken 26 (%54) hastada azalmıştı. Yeterli

kemik iliği megakaryositleri olan hastalar, azalmış olanlara göre daha

iyi bir yanıt oranına sahipti (%77’ye karşı %23, p<0,0001). Yeterli

megakaryositleri olan grup, tedaviye medyan 33 (aralık, 9-174) günde

yanıt verdi. Megakaryositleri azalmış hastalar, ortalama 55 (aralık, 30-

164) gün yanıt verdi (p=0,002). Hiçbir hastada ilaca bağlı yan etki

gözlenmedi.

Sonuç: Bu gerçek yaşam deneyimi, ELT’nin HKHN sonrası

trombositopeni için etkili ve güvenli bir tedavi seçeneği olduğunu

göstermektedir. ELT tedavisi öncesi kemik iliği megakaryositlerinin

yeterliliği tedaviye yanıtı etkileyen önemli bir faktördü.

Anahtar Sözcükler: Eltrombopag, Trombositopeni, Hematopoietik

kök hücre nakli, Trombosit iyileşmesi

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Zeynep Tuğba Güven, M.D., Erciyes University Faculty of Medicine,

Department of Hematology, Kayseri, Turkey

E-mail : drztkarabulutguven@gmail.com ORCID: orcid.org/0000-0003-1600-9731

Received/Geliş tarihi: December 8, 2021

Accepted/Kabul tarihi: March 11, 2022

103


Güven Z.T. et al: Eltrombopag for Post-transplant Thrombocytopenia

Turk J Hematol 2022;39:103-108

Introduction

Hematopoietic stem cell transplantation (HSCT) is an effective

treatment method used in the treatment of many malignant

and non-malignant diseases today. After transplantation,

engraftment is expected within weeks and blood parameters are

anticipated to return to normal levels; however, some patients

may suffer from cytopenia. The most frequently observed

cytopenia following allogeneic and autologous transplantation

is thrombocytopenia and it can be seen in up to 40% of patients

after transplantation [1,2,3]. Many underlying causes including

infections, side effects of drugs used, disease recurrence,

alloantibodies, insufficient graft function, and microangiopathy

can be identified [3,4,5]. Persistent thrombocytopenia after

HSCT is an important cause of mortality and morbidity

[1,2,6]. Thrombocytopenia may be severe in some patients and

may continue for a long time. However, there is no standard

approach for the treatment of these patients. Although it is

only a temporary fix, transfusion support is used often to treat

bleeding in such cases.

Eltrombopag (ELT) and romiplostim are thrombopoietin receptor

agonists (rhTPO) that increase platelet production and are used

in the treatment of idiopathic (immune) thrombocytopenic

purpura in adults and children [7,8,9,10]. In the literature,

it has been reported that ELT provides convenience with oral

use, and it is also used in the treatment of aplastic anemia,

low-risk myelodysplastic syndrome, and chemotherapyassociated

thrombocytopenia [11,12].

There are various reports on the use of ELT in the treatment

of post-transplant thrombocytopenia [13,14,15,16]. In this

study, we aim to share our experiences with ELT treatment in

48 patients with delayed platelet recovery (DPR) or secondary

failure of platelet recovery (SFPR) following HSCT.

Materials and Methods

Patients

The medical records of 634 patients who underwent HSCT at the

Erciyes University Bone Marrow Transplantation Center between

July 2017 and July 2021 were reviewed retrospectively. We

identified 48 patients treated with ELT for thrombocytopenia

after allogeneic or autologous transplantation. Successful

neutrophil engraftment was achieved in all cases. Patients

were classified into two groups as having experienced DPR or

SFPR. Patients with primary graft failure were not included in

the study and all patients were in remission. Signed written

consent forms were obtained from all participating patients

and the study was approved by the Ethics Committee of Erciyes

University (2020/156).

Definitions

Neutrophil engraftment was defined as an absolute neutrophil

count of ≥0.5x10 9 /L for 2 consecutive days. Platelet engraftment

was defined as a platelet count of ≥20x10 9 /L without transfusion

for 3 consecutive days. DPR was defined as the absence of

platelet engraftment on the 35 th day after transplantation

despite the occurrence of neutrophil engraftment [17]. SFPR

was defined as a platelet count of ≤20x10 9 /L after platelet

engraftment for more than 7 days [2,18]. Patients were excluded

if they had secondary causes of thrombocytopenia such as

drug-induced thrombocytopenia or viral infection-associated

thrombocytopenia. Liver function test results were normal for

all patients.

Bone marrow aspiration and biopsy were performed to evaluate

bone marrow cellularity just before the ELT treatment was

started. Megakaryocyte count was evaluated by bone marrow

aspirate smear and a count of ≥8/mm 2 was considered adequate

[13]. Platelet recovery after ELT was defined as a platelet

count above 50x10 9 /L for at least 7 days without the need for

transfusion.

Eltrombopag Treatment

While the initial dose of ELT was typically 25 mg or 50 mg, the dose

was increased by 25 mg per week based on the response status

and the maximum dose reached was 150 mg. The drug doses

given to the patients were adjusted according to their weekly

blood counts. When the platelet count exceeded 100x10 9 /L, the

dose was reduced by 25 mg weekly. Platelet transfusions were

performed according to institutional guidelines. Side effects

were graded according to Version 4.0 of the National Cancer

Institute Common Toxicity Criteria.

The efficacy of the treatment was determined by platelet count.

The primary outcome was a platelet count of ≥50x10 9 /L for 7

consecutive days without transfusion. Secondary outcomes

were the effect of bone marrow megakaryocyte count before

ELT treatment on platelet recovery and treatment-related side

effects.

Statistical Analysis

Continuous data conforming to normal distribution were

expressed as mean ± standard deviation, while continuous data

not conforming to normal distribution were expressed as median

and minimum-maximum and categorical data were expressed

as percentages (%). Categorical data were compared using

chi-square tests. The cumulative incidence of platelet recovery

was analyzed using Cox regression. Days without transfusion

from initiation of ELT to a platelet count of >50x10 9 /L were

compared between groups using the Wald chi-square test. Data

were analyzed with IBM SPSS Statistics 22.0 for Windows (IBM

Corp., Armonk, NY, USA). Values of p<0.05 were considered

significant.

Results

The clinical characteristics of the patients are summarized in

Table 1. Forty-eight HSCT recipients were included in this study.

104


Turk J Hematol 2022;39:103-108

Güven Z.T. et al: Eltrombopag for Post-transplant Thrombocytopenia

Thirty (62.5%) patients were evaluated as having experienced

DPR and 18 (37.5%) patients as having experienced SFPR. There

were 34 male patients, and the median age was 53 years (range:

21-69 years). Allogeneic stem cell transplantation was performed

for 28 (58%) patients and 18 of those patients were diagnosed

with acute myeloid leukemia. Myeloablative conditioning

regimens were used for all patients undergoing allogeneic

transplantation. Twenty-four patients of the allogeneic

transplantation group had a sibling HLA-matched donor, while

4 patients had a haploidentical donor. Peripheral blood was used

as a stem cell source in all cases. Cyclosporine and methotrexate

were used most frequently for graft-versus-host disease (GVHD)

prophylaxis, while post-transplant cyclophosphamide was

preferred in cases of haploidentical transplantations. Autologous

stem cell transplantation was performed for 20 (42%) patients

and 12 of those patients were diagnosed with non-Hodgkin

lymphoma. Neutrophil engraftment occurred in all patients

at a median of 17 days (range: 8-24 days). Grade II-IV GVHD

developed in 6 (12.5%) patients before ELT treatment. None of

the patients had a previous history of using rhTPO. Bone marrow

biopsy was performed for all patients before ELT treatment.

were 38% among patients with DPR and 50% among those

with SFPR for allogeneic transplantation, respectively. The

cumulative incidences of platelet recovery were 64% among

patients with DPR and 33% among those with SFPR for

autologous transplantation. Response rates were 55% and 43%

among patients who underwent autologous and allogeneic

transplantation, respectively. Patients with both DPR and

SFPR responded, but patients with DPR had a higher response

rate (50% vs. 44%). The median time to platelet recovery was

35 days (range: 9-174 days) and these patients had received

ELT for a median of 77 days (15-293 days). No recurrence of

thrombocytopenia was observed in any of these cases while ELT

was reduced. The median platelet count of the 23 responding

Outcomes of ELT treatment are listed in Table 2. ELT was started

at a median of 57 days after HSCT (range: 36-513 days). The

median platelet count before ELT treatment was 13x10 9 /L

(range: 3-20x10 9 /L). The starting dose of ELT was 50 mg per day

for most patients, with a maximum dose of 150 mg per day.

Twenty-three patients responded to treatment and the

cumulative incidence of successful platelet recovery was 48%

(Figure 1). The cumulative incidences of platelet recovery

Figure 1.Median platelet count before and after eltrombopag

treatment. The analysis included 23 responding patients

(p<0.0001).

Table 1. Clinical characteristics of the patients.

Characteristic Type of thrombocytopenia, n (%) All, n (%)

DPR (n=30) SFPR (n=18) (n=48)

Age, years, median (range) 54.5 (21-69) 50.5 (31-63) 53 (21-69)

Sex

Male

Female

Type of HSCT

Allo-HSCT

Auto-HSCT

Disease

AML

ALL

MM

HL

NHL

23 (77)

7 (23)

16 (53)

14 (47)

9 (30)

5 (17)

6 (20)

3 (10)

7 (23)

11 (61)

7 (39)

12 (67)

6 (33)

9 (50)

2 (11)

2 (11)

-

5 (28)

34 (71)

14 (29)

28 (58)

20 (42)

18 (37.5)

7 (14.5)

8 (17)

3 (6)

12 (25)

Neutrophil engraftment, day, median (range) 17 (10-24) 16 (8-23) 17 (8-24)

Grade II-IV GVHD 2 (7) 4 (22) 6 (12.5)

DPR: Delayed platelet recovery; SFPR: secondary failure of platelet recovery; HSCT: hematopoietic stem cell transplantation; Allo-HSCT: allogeneic HSCT; Auto-HSCT: autologous

HSCT; AML: acute myeloblastic leukemia; ALL: acute lymphoblastic leukemia; MM: multiple myeloma; HL: Hodgkin lymphoma; NHL: non-Hodgkin lymphoma; GVHD: graft-versushost

disease.

105


Güven Z.T. et al: Eltrombopag for Post-transplant Thrombocytopenia

Turk J Hematol 2022;39:103-108

patients was 12x10 9 /L (range: 5-19x10 9 /L) before treatment and

68x10 9 /L (52-266x10 9 /L) after treatment (p<0.0001; Figure 2).

Among the 25 non-responders, ELT was discontinued in 11 cases

due to death. It was continued for a median of 77 days among

non-responder patients (range: 10-232 days).

While the number of bone marrow megakaryocytes before

treatment was adequate in 22 (46%) cases, it was decreased

in 26 (54%) cases. Patients with adequate bone marrow

megakaryocytes had a better response rate than those without

(77% vs. 23%). This finding was significant (p<0.0001). The

group with adequate megakaryocytes responded to treatment

Figure 2. Cumulative incidence of platelet recovery (48%).

at a median of 33 days (range: 9-174 days), while patients with

decreased megakaryocytes responded at a median of 55 days

(30-164 days) (p=0.002; Table 3).

The treatment response was permanent in all patients and no

platelet count below 50x10 9 /L was observed even after ELT was

discontinued. The median follow-up time after discontinuation

of ELT treatment was calculated as 365 days (range: 61-921 days).

No drug-related side effects were observed in any patients. The

patients were fully compliant with treatment and none of the

patients had to discontinue the drug due to side effects.

Discussion

In this study, we have presented our single-center experience

with 48 patients treated with ELT for primary and secondary

platelet failure following HSCT. The cumulative incidence of

platelet recovery after treatment with ELT was 48%. These

patients, who needed continuous platelet transfusion, became

transfusion-free in a median of 35 days. Furthermore, after

discontinuation of ELT, platelet counts were maintained

permanently in all responding patients.

The use of ELT is effective in thrombocytopenia that develops

after autologous and allogeneic transplantation [13,19,20]. In

our study, response to ELT was observed in both the autologous

transplant and allogenic transplant groups (55% vs. 43%). In

Table 2. Outcomes of eltrombopag treatment.

Characteristics

Outcomes

Duration from transplantation to eltrombopag treatment, median (range), days 57 (36-513)

Starting dose of eltrombopag

25 mg daily, n (%)

50 mg daily, n (%)

Maximum dose of eltrombopag

75 mg daily, n (%)

100 mg daily, n (%)

125 mg daily, n (%)

150 mg daily, n (%)

6 (12.5)

42 (87.5)

3 (6)

17 (36)

4 (8)

24 (50)

Table 3. Bone marrow megakaryocytes of patients.

Achievement of platelet response

Yes, n (%)

No, n (%)

23 (48)

25 (52)

Days from starting eltrombopag to platelet response, median (range), days 35 (9-174)

Duration of treatment among patients with platelet response, median (range), days 77 (15-293)

Response

Adequate megakaryocytes

(n=22)

Positive response, n (%) 17 (77) 6 (23)

No response, n (%) 5 (23) 20 (77)

Days from starting eltrombopag to response,

median (range), days

Decreased megakaryocytes

(n=26)

p

<0.0001

33 (9-174) 55 (30-164) 0.002

106


Turk J Hematol 2022;39:103-108

Güven Z.T. et al: Eltrombopag for Post-transplant Thrombocytopenia

their study, Yuan et al. [21] used ELT therapy for 13 patients

with primary and secondary platelet failure. Response was

observed in 50% of the patients with primary failure and 71%

of those with secondary failure. In another study involving two

groups of patients who underwent autologous and allogeneic

transplantation, the response to ELT after transplantation was

100% and 61%, respectively. Hence, it was effective in cases

of both primary and secondary platelet failure [22]. In our

study, it was similarly observed that ELT was effective in both

primary and secondary platelet failure. There was more platelet

recovery in the group with primary failure (50% vs. 44%), but

that finding was not statistically significant.

Tanaka et al. [13] administered ELT for the treatment of

thrombocytopenia after allogeneic transplantation. Both

groups of patients with prolonged thrombocytopenia and

platelet recovery failure had positive results. Platelet recovery

was in line with the amount of bone marrow megakaryocytes

before treatment [13]. Similarly, patients with adequate

megakaryocyte counts prior to initiation of ELT therapy had

a higher platelet recovery rate than patients with decreased

megakaryocytes in the present study (77% vs. 23%, p<0.0001).

Based on these results, it can be thought that the number of

bone marrow megakaryocytes before treatment is a determining

factor in the response to rhTPO agonists. Patients with adequate

megakaryocytes responded to treatment in a shorter time than

patients with inadequate megakaryocytes (33 days vs. 55 days,

p=0.002). A normal megakaryocyte count allows the platelet

count to improve faster. In a study involving 13 patients,

ELT was used for thrombocytopenia following allogeneic

transplantation. The overall response rate was 62%. When

evaluated in terms of bone marrow reserve, the ELT response was

similar and sufficient in the group with lower megakaryocyte

counts [21]. It remains to be determined whether higher doses

of rhTPO agonists improve platelet recovery in patients with

reduced megakaryocyte counts. We used a maximum dose of

150 mg of ELT for our patients.

Study Limitations

Our study has several limitations; it was a retrospective study

and it included a small number of patients. In addition, the

patient group was heterogeneous. Although we investigated

other causes of thrombocytopenia for all patients, it was not

always possible to exclude them completely. However, we think

that our study nevertheless supports the safety and efficacy of

ELT in the treatment of thrombocytopenia following HSCT.

Conclusion

The results that we have presented here demonstrate that ELT

is an effective and safe treatment option for thrombocytopenia

following HSCT. However, prospective randomized studies are

needed to demonstrate the efficacy and optimal dose of ELT

in the treatment of post-transplant thrombocytopenia. Patients

with normal megakaryocyte counts responded better and

faster to ELT. Adequacy of bone marrow megakaryocytes before

ELT treatment was an important factor affecting treatment

response.

Ethics

Ethics Committee Approval: Signed written consent forms

were obtained from all participating patients and the study

was approved by the Ethics Committee of Erciyes University

(2020/156).

Informed Consent: Signed written consent forms were obtained.

Authorship Contributions

Surgical and Medical Practices: Z.T.G., S.Ç., B.E., M.Ç., A.Ü., L.K.;

Concept: Z.T.G., L.K., Design: Z.T.G., L.K.; Data Collection or

Processing: Z.T.G., S.Ç., B.E.; Analysis or Interpretation: Z.T.G.,

S.Ç., B.E., M.Ç.; Literature Search: Z.T.G., S.Ç., B.E., M.Ç., A.Ü., L.K.;

Writing: Z.T.G., S.Ç.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Kuzmina Z, Eder S, Böhm A, Pernicka E, Vormittag L, Kalhs P, Petkov V,

Stary G, Nepp J, Knobler R. Significantly worse survival of patients with

NIH-defined chronic graft-versus-host disease and thrombocytopenia

or progressive onset type: results of a prospective study. Leukemia

2012;26:746-756.

2. Bruno B, Gooley T, Sullivan KM, Davis C, Bensinger WI, Storb R, Nash

RA. Secondary failure of platelet recovery after hematopoietic stem cell

transplantation. Biol Blood Marrow Transplant 2001;7:154-162.

3. Yamazaki R, Kuwana M, Mori T, Okazaki Y, Kawakami Y, Ikeda Y, Okamoto

S. Prolonged thrombocytopenia after allogeneic hematopoietic stem

cell transplantation: associations with impaired platelet production and

increased platelet turnover. Bone Marrow Transplant 2006;38:377-384.

4. Dominietto A, Raiola AM, Van Lint MT, Lamparelli T, Gualandi F, Berisso

G, Bregante S, Frassoni F, Casarino L, Verdiani S. Factors influencing

haematological recovery after allogeneic haemopoietic stem cell

transplants: graft‐versus‐host disease, donor type, cytomegalovirus

infections and cell dose. Br J Haematol 2001;112:219-227.

5. Culligan D. Clinical bone marrow and blood stem cell transplantation. Br J

Haematol 2001;112:254-255.

6. Bolwell B, Pohlman B, Sobecks R, Andresen S, Brown S, Rybicki L, Wentling

V, Kalaycio M. Prognostic importance of the platelet count 100 days post

allogeneic bone marrow transplant. Bone Marrow Transplant 2004;33:419-

423.

7. Bussel JB, Cheng G, Saleh MN, Psaila B, Kovaleva L, Meddeb B, Kloczko J,

Hassani H, Mayer B, Stone NL. Eltrombopag for the treatment of chronic

idiopathic thrombocytopenic purpura. N Engl J Med 2007;357:2237-2247.

107


Güven Z.T. et al: Eltrombopag for Post-transplant Thrombocytopenia

Turk J Hematol 2022;39:103-108

8. Bussel JB, Kuter DJ, Pullarkat V, Lyons RM, Guo M, Nichol JL. Safety and

efficacy of long-term treatment with romiplostim in thrombocytopenic

patients with chronic ITP. Blood 2009;113:2161-2171.

9. Bussel JB, de Miguel PG, Despotovic JM, Grainger JD, Sevilla J, Blanchette

VS, Krishnamurti L, Connor P, David M, Boayue KB. Eltrombopag

for the treatment of children with persistent and chronic immune

thrombocytopenia (PETIT): a randomised, multicentre, placebo-controlled

study. Lancet Haematol 2015;2:e315-e325.

10. Mishra K, Pramanik S, Jandial A, Sahu KK, Sandal R, Ahuja A, Yanamandra

U, Kumar R, Kapoor R, Verma T. Real-world experience of eltrombopag in

immune thrombocytopenia. Am J Blood Res 2020;10:240.

11. Townsley DM, Scheinberg P, Winkler T, Desmond R, Dumitriu B, Rios O,

Weinstein B, Valdez J, Lotter J, Feng X. Eltrombopag added to standard

immunosuppression for aplastic anemia. N Engl J Med 2017;376:1540-

1550.

12. Olnes MJ, Scheinberg P, Calvo KR, Desmond R, Tang Y, Dumitriu B, Parikh

AR, Soto S, Biancotto A, Feng X. Eltrombopag and improved hematopoiesis

in refractory aplastic anemia. N Engl J Med 2012;367:11-19.

13. Tanaka T, Inamoto Y, Yamashita T, Fuji S, Okinaka K, Kurosawa S, Kim SW,

Tanosaki R, Fukuda T. Eltrombopag for treatment of thrombocytopenia

after allogeneic hematopoietic cell transplantation. Biol Blood Marrow

Transplant 2016;22:919-924.

14. Dyba J, Tinmouth A, Bredeson C, Matthews J, Allan D. Eltrombopag after

allogeneic haematopoietic cell transplantation in a case of poor graft

function and systematic review of the literature. Transfus Med 2016;26:202-

207.

15. Bosch‐Vilaseca A, García‐Cadenas I, Roldán E, Novelli S, Barba P, Esquirol

A, Valcárcel D, Martino R, Sierra J. Usefulness of thrombopoietin receptor

agonists for persistent clinically relevant thrombocytopenia after allogeneic

stem cell transplantation. Eur J Haematol 2018;101:407-414.

16. Marano L, Marotta S, Cacace F, Frieri C, Simeone L, Trastulli F, Vitiello S,

Cardano F, Pane F, Risitano AM. Eltrombopag for prolonged post-transplant

hyporigenerative cytopenias. Blood 2018;132:5711.

17. Nash RA, Kurzrock R, DiPersio J, Vose J, Linker C, Maharaj D, Nademanee

AP, Negrin R, Nimer S, Shulman H. A phase I trial of recombinant

human thrombopoietin in patients with delayed platelet recovery after

hematopoietic stem cell transplantation. Biol Blood Marrow Transplant

2000;6:25-34.

18. Akahoshi Y, Kanda J, Gomyo A, Hayakawa J, Komiya Y, Harada N, Kameda

K, Ugai T, Wada H, Ishihara Y. Risk factors and impact of secondary failure

of platelet recovery after allogeneic stem cell transplantation. Biol Blood

Marrow Transplant 2016;22:1678-1683.

19. Reid R, Bennett JM, Becker M, Chen Y, Milner L, Phillips GL 2nd, Liesveld J. Use

of eltrombopag, a thrombopoietin receptor agonist, in post‐transplantation

thrombocytopenia. Am J Hematol 2012;87:743-745.

20. Raut SS, Shah SA, Sharanangat VV, Shah KM, Patel KA, Anand AS, Talati

SS, Panchal HP, Patel AA, Parikh SK. Safety and efficacy of eltrombopag

in post-hematopoietic stem cell transplantation (HSCT) thrombocytopenia.

Indian J Hematol Blood Transfus 2015;31:413-415.

21. Yuan C, Boyd AM, Nelson J, Patel RD, Varela JC, Goldstein SC, Ahmad S, Zhu

X, Mori S. Eltrombopag for treating thrombocytopenia after allogeneic stem

cell transplantation. Biol Blood Marrow Transplant 2019;25:1320-1324.

22. Mori S, Patel RD, Boyd A, Simon S, Nelson J, Goldstein SC. Eltrombopag

treatment for primary and secondary thrombocytopenia post allogeneic

and autologous stem cell transplantation is effective and safe. Biol Blood

Marrow Transplant 2018;24:S343-S344.

108


RESEARCH ARTICLE

DOI: 10.4274/tjh.galenos.2021.2021.0325

Turk J Hematol 2022;39:109-116

Assessment of Bone Marrow Biopsy and Cytogenetic Findings in

Patients with Multiple Myeloma

Multipl Myelomlu Hastalarda Kemik İliği Biyopsisi ve Sitogenetik Bulguların

Değerlendirilmesi

Ahmet Şeyhanlı 1 , Boran Yavuz 2 , Zehra Akşit 3 , Zeynep Yüce 4 , Sermin Özkal 5 , Oğuz Altungöz 4 , Fatih Demirkan 2 ,

İnci Alacacıoğlu 2 , Güner Hayri Özsan 2

1Sivas Numune Hospital, Clinic of Hematology, Sivas, Turkey

2Dokuz Eylül University Faculty of Medicine, Department of Hematology, İzmir, Turkey

3Dokuz Eylül University Faculty of Medicine, Department of Internal Medicine, İzmir, Turkey

4Dokuz Eylül University Faculty of Medicine, Department of Medical Biology, İzmir, Turkey

5Dokuz Eylül University Faculty of Medicine, Department of Pathology, İzmir, Turkey

Abstract

Objective: Multiple myeloma (MM) is a malignant condition

characterized by the accumulation of malignant plasma cells.

Although MM remains incurable, the survival of MM patients has

improved considerably due to the application of autologous stem cell

transplantation, novel agents, and advanced treatment strategies. This

study aimed to determine the cytogenetic characterization and bone

marrow (BM) features of Turkish patients with MM.

Materials and Methods: Eighty-five MM patients were admitted to

Dokuz Eylül University Hospital in Turkey. BM samples of these MM

patients were subjected to cytogenetic analyses at diagnosis and

during therapy as a part of therapeutical and clinical evaluation.

A complete cytogenetic study was performed using the G-banding

technique. Fluorescence in situ hybridization (FISH) analysis was

performed using cytoplasmic immunoglobulin. The degree of BM

fibrosis was determined using reticulin histochemical staining. We

determined the percentage of BM plasma cells based on the extent

of CD38 staining.

Results: Eighty-five MM patients were retrospectively identified

between 2015 and 2021. The median age was 63 (38-90) years. Of

the 85 patients, 60 (70.6%) were male and 25 (29.4%) were female.

Seventy-two (84.7%) cases had BM fibrosis at the time of diagnosis.

The most common was grade 2 fibrosis, recorded in 35 cases (41.2%).

About 72.9% of the patients showed more than 50% plasma cells.

FISH analysis indicated the presence of abnormal chromosomes in

37% (32/85) of the patients. The most frequent abnormality was

Immunoglobulin heavy-chain (IGH) translocation (21.3%).

Conclusion: Subgroup analysis of IGH mutations is crucial in the

identification of high-risk MM patients. We believe that our study

will contribute to the determination of BM biopsy and cytogenetic

features of MM patients in our country.

Keywords: Multiple myeloma, Bone marrow biopsy, Cytogenetic

analysis, Fluorescence in situ hybridization

Öz

Amaç: Multipl myelom (MM), malign plazma hücrelerinin birikmesi

ile karakterize malign bir durumdur. MM tam kür sağlamanan hastalık

olmasa da, uygulanan otolog kök hücre nakli ve yeni ilaçlar ve yeni

tedavi stratejileri nedeniyle MM hastalarının sağkalımı önemli

ölçüde gelişmiştir. Bu çalışmada MM’li Türk hastalarda sitogenetik

karakterizasyon ve kemik iliği (Kİ) özelliklerinin belirlenmesi

amaçlanmıştır.

Gereç ve Yöntemler: Türkiye’deki Dokuz Eylül Üniversite

Hastanesi’ndeki 85 MM hastasını kaydettik. Bu MM hastalarının kemik

iliği örneklerinde, tedavi ve klinik değerlendirmenin bir parçası olarak

tanı anında ve tedavi süresince sitogenetik analiz yapıldı. G-bantlama

tekniği kullanılarak tam bir sitogenetik çalışma gerçekleştirildi.

Fazlar arası Floresan in situ hibridizasyon (FISH) analizi, sitoplazmik

immünoglobulin ile yapıldı. Kemik iliği fibrozunun derecesi, retikülinin

histokimyasal boyası kullanılarak belirlendi. Kİ plazma hücrelerinin

yüzdesi, CD38 boyamasınına göre belirlendi.

Bulgular: 2015 ve 2021 yılları arasında geriye dönük olarak 85 MM

hastası belirlendi. Ortanca yaş 63 (38-90) yıldı. Seksen beş hastanın

60’ı (%70,6) erkek, 25’i (%29,4) kadındı. Tanı anında kemik iliği

fibrozu 72 olguda (%84,7) mevcuttu. En sık 35 hastada (%41,2) derece

2 fibrozis görüldü. Hastaların %72,9’unda plazma hücre yüzdeleri

%50’den fazlaydı. FISH analizi sonuçlarında kromozom anomalilerinin

varlığı %37 (32/85) oranındadır. En sık kromozom görülen anormallik

IGH translokasyonuydu (%21,3).

Sonuç: IGH mutasyonlarının alt grup analizi, yüksek riskli MM

hastalarının belirlenmesinde kritik öneme sahiptir. Çalışmamızın

ülkemizdeki MM hastalarının Kİ biyopsisi ve sitogenetik özelliklerinin

belirlenmesine katkı sağlayacağına inanıyoruz.

Anahtar Sözcükler: Multipl myelom, Kemik iliği biyopsisi, Sitogenetik

analiz, Floresan in situ hibridizasyon

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Ahmet Şeyhanlı, M.D., Sivas Numune Hospital, Clinic of Hematology,

Sivas, Turkey

E-mail : ahmet8563@yahoo.com ORCID: orcid.org/0000-0001-6082-2995

Received/Geliş tarihi: May 22, 2021

Accepted/Kabul tarihi: November 25, 2021

109


Şeyhanlı A. et al: Genetic and Bone Marrow Features in Multiple Myeloma

Turk J Hematol 2022;39:109-116

Introduction

Multiple myeloma (MM) is a malignant condition characterized

by the accumulation of malignant plasma cells. It is the second

most common hematological malignancy that develops in

the bone marrow (BM) [1]. Although MM remains incurable,

the survival of MM patients has improved considerably due

to the application of autologous stem cell transplantation

(ASCT), novel agents, and advanced treatment strategies.

However, this development has not been uniform. The existing

heterogeneity is dependent on patient-specific factors such

as age, comorbidities, and disease-related factors, including

cytogenetic and molecular features, BM fibrosis (BMF), and the

plasma cells in the BM. Several risk classifications have been

made for personalized therapeutic approaches. A clinical staging

system for MM was first developed by Durie and Salmon [2]

in 1975. The International Staging System (ISS) score was then

defined by Greipp et al. [3] in 2005 based on two parameters:

serum β2-microglobulin level and serum albumin. Avet-Loiseau

et al. [4] and Cavo-Rosinol et al. [5] combined cytogenetics with

the ISS to improve risk stratification. Several techniques are

available to detect genetic abnormalities in MM. Karyotyping is

applied to detect cytogenetic abnormalities and abnormalities

dependent on the proliferative index of malignant plasma

cells but provides limited information in vitro due to the low

proliferative ability of malignant plasma cells [6]. Kishimoto et al.

[7]reported that abnormal karyotypes are seen in approximately

30%-50% of MM cases. The gold-standard approach for the

detection of genomic abnormalities in MM is fluorescence in

situ hybridization (FISH), which has been implemented and

validated by various cytogenetic laboratories around the globe

[8]. The identification of high-risk subtypes of myeloma is vital.

According to previous works, high-risk MM patients showed

a 20%-50% reduction in overall survival (OS) compared to

standard-risk patient groups when undergoing induction with

conventional chemotherapy followed by ASCT. The present

study aimed to determine the cytogenetic characters and BM

features of Turkish patients with MM.

Materials and Methods

From January 2015 to January 2021, 85 MM patients were

admitted to Dokuz Eylül University Hospital in Turkey. BM samples

of these MM patients were subjected to cytogenetic analyses

at diagnosis and during therapy as a part of therapeutical and

clinical evaluation. Conventional cytogenetic analysis was

performed for all samples at presentation and after a short-term

culture period of 24/72 h. Cells were treated with colcemid and

harvested for 20 min. Karyotypes were then analyzed using the

standard G-banding technique. A minimum of 20 metaphases

per case were analyzed. The karyotypes were reported according

to the 2016 International System for Human Cytogenetic

Nomenclature. An interphase FISH study was performed on BM

samples according to the relevant manufacturer’s hybridization

and post-hybridization stringency conditions with minimal

modifications. Dual-color CytoCell FISH probes specific to loci

13q14.3 (LPH006), CKS1B (1q21-22)/CDKN2C (1p32.3) (LPH039),

IGH (14q32.33) (Break-apart, LPH 014), P53 (17p13.1)/ATM

(11q32) (LPH 052), and MLL (11q23.3) (Break-apart, LPH 013)

were used. Threshold values of copy number gain, deletion,

and break-apart positivity were 7%, 7%, and 10%, respectively.

FISH slides were analyzed with a motorized Olympus BX61

fluorescence microscope equipped with 4′,6-diamidino-

2-phenylindole, fluorescein isothiocyanate, rhodamine, dual,

and triple band-pass filters (Chroma, Bellows Falls, VT, USA). Two

independent observers analyzed the slides blindly, enumerating

at least 200 optimally hybridized nuclei. The degree of BMF

was detected using reticulin histochemical staining. The level

of fibrosis as determined by a pathologist was scored according

to the guidelines of the World Health Organization [grade 0:

no fibrosis, grade 1 (mild): low (fine reticulin network), grade 2

(moderate): intermediate (multifocal or diffuse non-confluent

fibrosis), grade 3 (severe): high (marked and diffuse fibrosis)].

The percentage of BM plasma cells was determined based on

the extent of CD38 staining and categorized as Group 1 (<20%),

Group 2 (20%-50%), or Group 3 (>50% plasma cells). The ISS

was used based on the combination of serum β2-microglobulin

and serum albumin.

Statistical Analysis

IBM SPSS Statistics 24.0 for Windows (IBM Corp., Armonk, NY,

USA) was used for statistical analysis. Numerical variables are

presented as median or mean, while categorical variables are

given as number (n) and percentage (%). The OS curves were

estimated using the Kaplan-Meier method whereby OS was

defined as the time from the diagnosis of MM to death from any

cause. The confidence interval was determined as 95% in the

analyses and values of p<0.05 were considered to be statistically

significant.

Results

Eighty-five MM patients were retrospectively identified between

2015 and 2021. The median age was 63 (38-90) years and 56.5%

of the patients were under 65 years old. Of the 85 patients, 60

(70.6%) were male and 25 (29.4%) were female. Bone lesions

were detected in 57.6% of these cases, anemia in 42.4%,

impaired renal function in 27.1%, and hypercalcemia in 18.8%.

Most patients were in ISS stage III. More than half of the patients

received proteasome inhibitor-based therapy for induction.

Information about the induction therapy of five patients

could not be obtained. Further details about the demographic,

laboratory, and clinical characteristics are presented in Table 1.

110


Turk J Hematol 2022;39:109-116

Şeyhanlı A. et al: Genetic and Bone Marrow Features in Multiple Myeloma

Table 1. Demographic, laboratory, and clinical characteristics

of multiple myeloma (MM) patients.

Characteristics n (%)

Age, median, years (range) 63 years (38-90)

<65 years 48 (56.5)

≥65 years 37 (43.5)

Sex

Female 25 (29.4)

Male 60 (70.6)

Immunoglobulin (Ig) isotype

IgG 48 (56.5)

IgA 13 (15.3)

Light chain MM 24 (28.2)

Type of light chain

Kappa 54 (63.5)

Lambda 31 (26.5)

ISS stage

I 21 (24.7)

II 23 (27.1)

III 41 (48.2)

Anemia

Hemoglobin >10 g/dL 49 (57.6)

Hemoglobin <10 g/dL 36 (42.4)

Renal impairment

Creatinine ≤2 mg/dL 62 (72.9)

Creatinine >2 mg/dL 23 (27.1)

Hypercalcemia

Serum calcium <11 mg/dL 69 (81.2)

Serum calcium >11 mg/dL 16 (18.8)

Bone lesions

Absent 36 (42.4)

Present 49 (57.6)

High ESR level (>15 mm/h)

Absent 26 (30.5)

Present 52 (61.1)

Not available 7 (8.4)

High LDH level (>250 U/L)

Absent 60 (70.6)

Present 25 (29.4)

High ferritin level (>335 ng/mL)

Absent 51 (60.0)

Present 17 (20.0)

Not available 17 (20.0)

Treatment type used for induction

VAD 4 (4.70)

Proteasome inhibitor 54 (63.5)

Immunomodulatory drug 3 (3.52)

Both proteasome inhibitor and

immunomodulatory drug

19 (22.3)

Not available 5 (5.88)

Total 85 (100)

ISS: International Staging System; ESR: erythrocyte sedimentation rate; LDH: lactate

dehydrogenase; VAD: vincristine, doxorubicin, and dexamethasone.

BMF was observed at the time of diagnosis in 72 cases (84.7%).

The most common finding was grade 2 fibrosis in 35 patients

(41.2%). In 72.9% of the patients, a plasma cell percentage of

over 50% was observed, and the most common type of plasma

cell infiltration was the diffuse involvement pattern (50.6%).

Based on FISH analysis, 37% (32/85) of cases showed the presence

of chromosomal anomalies. The most frequent abnormality was

the IGH translocation (21.3%). Based on karyotype analysis,

54.4% (37/68) of the patients had normal karyotypes while

5.8% (4/68) had abnormal karyotypes. Table 2 presents plasma

cell percentages, plasma cell infiltration patterns, and fibrosis

results from BM biopsy samples. Detailed cytogenetic results of

these MM patients are presented in Table 3.

In this study, the median OS of the entire population was 29.4

months (Figure 1a). Del(17p13)-negative patients had longer OS

compared to del(17p13)-positive patients (32.4 months vs. 11.1

months, p=0.015) (Figure 1b).

IGH-negative and del(13q)-negative patients had longer OS

compared to IGH-positive and del(13q)-positive patients.

However, this was not statistically significant (p=0.594 and

p= 0.094, respectively). A lower percentage of plasma cells in

the BM was associated with better OS than higher plasma cell

percentages. The median OS was 42.6 months in patients with

0%-19% plasma cells, 33.5 months in patients with 20%-49%

plasma cells, and 28.4 months in patients with 50%-100%

plasma cells. However, this observation was not statistically

significant (p=0.969). The results of multivariate analysis

adjusting for the del(17p13) mutation, IGH mutation, del(13)

mutation, BMF, plasma cell infiltration pattern, and plasma cell

percentage are shown in Table 4.

Table 2. Analysis of plasma cell percentage, plasma cell

infiltration pattern, and fibrosis in bone marrow biopsy

samples from patients with multiple myeloma at diagnosis.

Bone marrow plasma cell percentage (%) n (%)

0-19 5 (5.9)

20-49 62 (72.9)

50-100

Pattern of plasma cell infiltration 33 (38.8)

Interstitial 9 (10.6)

Nodular

Diffuse 43 (50.6)

Bone marrow fibrosis

MF-0 13 (15.3)

MF-1 35 (41.2)

MF-2 9 (10.6)

MF-3

Total 85 (100)

MF: Marrow fibrosis.

111


Şeyhanlı A. et al: Genetic and Bone Marrow Features in Multiple Myeloma

Turk J Hematol 2022;39:109-116

Figure 1. a) Kaplan-Meier curve of overall survival (OS) for the entire study cohort (n=85). The median OS was 29.4 months. b) Del(17p13)-

negative patients had longer OS compared to del(17p13)-positive patients (32.4 months vs. 11.1 months, p=0.015).

Table 3. Detailed cytogenetic results of multiple myeloma patients.

Characteristics Information available, n Positive patients, n (%)

Del(17p13) mutation 85 6 (7.1)

Del(13q14) mutation 49 9 (18.4)

IGH translocations 80 17 (21.3)

CKS1B mutation 1 0 (0.00)

MLL mutation 15 0 (0.00)

Karyotyping 68

Normal 37 (54.4)

Hyperdiploid 2 (2.9)

Non-hyperdiploid 2 (2.9)

Non-diagnostic 27 (39.7)

Discussion

MM remains an incurable malignancy with varying clinical and

prognostic differences depending on various factors including

age, comorbidity, cytogenetic and genomic features, and tumor

microenvironment characteristics in the bone marrow. The

disease incidence ranges from 5 to 7 per 100,000 and increases

with age. The median age at diagnosis of MM is 69 years. Fewer

than 14% of patients are younger than 55 years. MM is seen

more often in men and African Americans. The etiological

reasons behind these differences are not sufficiently understood.

Survival among patients with MM ranges from 1 year to more

than 10 years. Median survival in unselected patients with MM

is 3 years. The 5-year relative survival rate is 46.6% [9]. In our

study, the median age was 63 and male patients constituted

70.6% of the total. These findings are slightly different from

those published in the literature, possibly due to environmental

and genetic factors.

Understanding the alterations in the BM microenvironment

and the molecular pathways related to these changes is

crucial for the further improvement of MM treatment and

the outcomes of these patients. Sailer et al. [10] demonstrated

that characteristics of the tumor microenvironment such as

plasma cell differentiation, volume of plasma cell infiltration,

and infiltration pattern have predictive value. However, the

prognostic efficacy of BMF at the time of diagnosis is still

controversial [11,12]. Babarović et al. [13] reported that the

5-year survival rates of patients with increased BMF subjected

to post-treatment BM biopsy were significantly shorter than

112


Turk J Hematol 2022;39:109-116

Şeyhanlı A. et al: Genetic and Bone Marrow Features in Multiple Myeloma

Table 4. Multivariable Cox regression analysis for overall survival (OS).

Characteristics Median OS (months) Hazard ratio 95% CI p

Del(17p13) 0.015

Del(17p13)-negative 32.4 1

Del(17p13)-positive 11.1 3.45 1.18-10.0

IGH translocations 0.594

IGH-negative 28.9 1

IGH-positive 22.8 1.35 0.44-4.11

Del(13q) mutation 0.094

Del(13q)-negative 28.4 1

Del(13q)-positive 23.3 1.88 0.89-3.96

Bone marrow fibrosis 0.938

MF-0 34.4 1

MF-1 33.0 0.90 0.35-2.29

MF-2 27.4 1.15 0.46-2.87

MF-3 33.4 1.08 0.31-3.72

Pattern of plasma cell infiltration 0.466

Interstitial 33.4 1

Nodular 23.3 1.80 0.69-473

Diffuse 28.4 1.12 0.55-2.27

Bone marrow plasma cell percentage (%)

0-19 42.6 1 0.969

20-49 33.5 0.86 0.23-3.31

50-100 28.4 0.97 0.29-3.26

CI: Confidence interval; MF: marrow fibrosis; IGH: immunoglobulin heavy chain.

those of patients without fibrosis. Thus, the assessment of BMF

was concluded to have prognostic value in the follow-up of MM

patients [13]. A recent study by Paul et al. [14] showed that the

median progression-free survival in patients without BMF was

30.2 months, while in those with BMF, it was 21 months. On

the other hand, median OS was 61.2 months and 45.1 months

among patients without and with BMF, respectively. These

findings were statistically significant [14]. Our results suggested

that the pattern of plasma cell infiltration and the degree of

fibrosis at diagnosis did not significantly influence survival

times, possibly due to the relatively small groups in this study.

Several predictive models like the Durie and Salmon [2] staging

system and the ISS have been used to categorize MM patients

into risk groups. The Durie and Salmon [2] staging system was

the first such system devised for MM and it has been widely

used for several decades. Later proposed by Greipp et al. [3], the

ISS became more widely used. The ISS is based on the prognostic

factors of serum β2-microglobulin and serum albumin. For

ISS stages I, II, and III, the median survival of MM patients

was determined to be 62 months, 44 months, and 29 months,

respectively [3]. A major limitation of the ISS classification is

that it does not consider patient-specific factors. Approximately

half of our patients were in ISS stage III and their median OS was

28.4 months. Our results were thus consistent with the study

of Greipp et al. [3]. Chromosomal abnormalities detected by

FISH were combined with the ISS to improve risk stratification

[4,5]. Because karyotypes are highly sensitive in the detection

113


Şeyhanlı A. et al: Genetic and Bone Marrow Features in Multiple Myeloma

Turk J Hematol 2022;39:109-116

of numerical chromosomal abnormalities, patients can be

classified into specific categories such as normal, hyperdiploid,

or non-hyperdiploid (including hypodiploid, pseudodiploid,

and near-tetraploid) karyotypes [15]. Among non-hyperdiploid

cases of MM, the hypodiploid subtype is associated with

poor prognosis [16,17]. In our study, abnormal karyotypes

were detected in 4 of 68 (5.8%) patients. Among these cases,

non-hyperdiploid abnormalities were observed in two patients.

However, chromosomal abnormalities could not be evaluated in

39.7% of the total cases because of the absence of metaphase

cells. FISH does not require metaphase cells before analysis

and it is accepted as the gold-standard test for the detection

of genomic abnormalities in MM, being applied and verified

by cytogenetics laboratories around the globe [8]. Further

studies are underway for identifying the optimal risk-based

classification and treatment for MM and especially for the

identification of high-risk MM patients. The International

Myeloma Working Group provided a consensus decision stating

that FISH panel testing should be performed for at least t(4;14),

t(14;16), and del(17p13) to define patients with high-risk disease

[18]. In addition to that recommendation, Boyd et al. [19]

suggested that FISH panel tests for +1q21 and t(14;20) should

be performed in MM risk classification. Deletion of chromosome

17p13 increases the immortalization of tumor cells by inhibiting

apoptosis [20]. Del(17p13) is observed in about 10% of MM

cases and has been associated with impaired OS [21,22,23,24].

In our study, the incidence rate of the del(17p13) mutation was

7.1% and del(17p13)-negative patients had significantly longer

OS compared to del(17p13)-positive patients (32.4 months vs.

11.1 months, respectively). Previously, del(13q) was in the poor

prognosis group, but it is now excluded from risk stratification

as it was detected in almost 50% of myeloma patients and is

thought to be secondary to the close association with t(4;14),

del(17p) [19,25,26]. In this study, del(13q) was detected in 9 of

49 cases (18.4%) and a significant adverse effect of del(13q) on

OS was not observed (p=0.094).

Chromosome 14q32 translocations (heavy chain IGH

translocations) in MM also increase dysregulated proliferation

and inhibit differentiation. Bergsagel and Kuehl [27] reported an

incidence of IGH translocations of 60%-65% in intramedullary

MM. However, in our study, the incidence of IGH mutations

was lower (21.3%). Over 20 different IGH translocations

have been revealed, among which IGH translocations t(4;14),

t(14;16), and t(14;20) are associated with poor prognosis in MM

patients [28]. The use of proteasome inhibitor-based induction

is recommended to improve the prognosis of patients with

t (4;14) but not those with del(17p13) [29]. Unfortunately, our

study does not include subgroup analysis of IGH mutations

that involve several recurrent translocations. Recently, gene

expression profiling methods, such as sequencing comparative

genomic hybridization (CGH), single nucleotide polymorphism

sequencing CGH, gene expression profiling, and RNA sequencing

have been introduced to identify patients with particularly

high-risk MM [30,31,32]. However, this is not feasible in routine

practice due to the high costs.

Study Limitations

The present study has a few limitations. It was a retrospective

study based on single-center data. Possibly because of the small

sample size, we did not observe a significant adverse effect of

IGH mutation on OS. Since our study did not include subgroups

of IGH mutations, high-risk patients could not be determined

clearly.

Conclusion

To the best of our knowledge, this study is the first of its

kind conducted in Turkey to include genetic results. This is

especially crucial in the identification of high-risk myeloma

subtypes. The subgroup analysis of IGH mutations is critical in

identifying high-risk MM patients. IGH subgroup analyses have

been conducted recently in our center. The present study will

contribute to determining BM biopsy and cytogenetic features

of MM patients in our country.

Ethics

Ethics Committee Approval: This study was approved by the

Ethics Committee of Dokuz Eylül University (2021/10-25).

Informed Consent: Retrospective study.

Authorship Contributions

Concept: A.Ş., B.Y., Z.A., Z.Y., S.Ö., O.A., F.D., İ.A., G.H.Ö.; Design:

A.Ş., B.Y., Z.A., Z.Y., S.Ö., O.A., F.D., İ.A., G.H.Ö.; Data Collection or

Processing: A.Ş., B.Y., Z.A., Z.Y., S.Ö., O.A., F.D., İ.A., G.H.Ö.; Analysis

or Interpretation: A.Ş., B.Y., Z.A., Z.Y., S.Ö., O.A., F.D., İ.A., G.H.Ö.;

Literature Search: A.Ş., B.Y., Z.A., Z.Y., S.Ö., O.A., F.D., İ.A., G.H.Ö.;

Writing: A.Ş., B.Y., Z.A., Z.Y., S.Ö., O.A., F.D., İ.A., G.H.Ö.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Kyle RA, Rajkumar SV. ASH 50th anniversary review. Blood 2008;111:2962-

2972.

2. Durie BG, Salmon SE. A clinical staging system for multiple myeloma

correlation of measured myeloma cell mass with presenting clinical

features, response to treatment, and survival. Cancer 1975;36:842-854.

3. Greipp PR, San Miguel J, Durie BG, Crowley JJ, Barlogie B, Bladé J,

Boccadoro M, Child JA, Avet-Loiseau H, Kyle RA, Lahuerta JJ, Ludwig H,

Morgan G, Powles R, Shimizu K, Shustik C, Sonneveld P, Tosi P, Turesson I,

Westin J. International staging system for multiple myeloma. J Clin Oncol

2005;23:3412-3420.

114


Turk J Hematol 2022;39:109-116

Şeyhanlı A. et al: Genetic and Bone Marrow Features in Multiple Myeloma

4. Avet-Loiseau H, Durie BG, Cavo M, Attal M, Gutierrez N, Haessler J,

Goldschmidt H, Hajek R, Lee JH, Sezer O, Barlogie B, Crowley J, Fonseca R,

Testoni N, Ross F, Rajkumar SV, Sonneveld P, Lahuerta J, Moreau P, Morgan

G; International Myeloma Working Group. Combining fluorescent in situ

hybridization data with ISS staging improves risk assessment in myeloma:

an International Myeloma Working Group collaborative project. Leukemia

2013;27:711-717.

5. Cavo M, Salwender H, Rosiñol L, Moreau P, Petrucci MT, Blau IW, Bladé J,

Attal M, Patriarca F, Weisel K, San Miguel JF, Avet-Loiseau H, Testoni N,

Pfreundschuh M, Lahuerta JJ, Facon T, Pantani L, Scheid C, Gutierrez N,

Marit G, Palumbo A, Martin ML, Caillot D, Goldschmidt H. Double vs single

autologous stem cell transplantation after bortezomib-based induction

regimens for multiple myeloma: an integrated analysis of patient-level data

from phase European III studies. Blood 2013;122:767.

6. Avet-Loiseau H, Hulin C, Campion L, Rodon P, Marit G, Attal M, Royer B,

Dib M, Voillat L, Bouscary D, Caillot D, Wetterwald M, Pegourie B, Lepeu

G, Corront B, Karlin L, Stoppa AM, Fuzibet JG, Delbrel X, Guilhot F, Kolb B,

Decaux O, Lamy T, Garderet L, Allangba O, Lifermann F, Anglaret B, Moreau

P, Harousseau JL, Facon T. Chromosomal abnormalities are major prognostic

factors in elderly patients with multiple myeloma: the Intergroupe

Francophone du Myélome experience. J Clin Oncol 2013;31:2806-2809.

7. Kishimoto RK, Freitas SLVVd, Ratis CA, Borri D, Sitnik R, Velloso EDRP.

Validation of interphase fluorescence in situ hybridization (iFISH) for

multiple myeloma using CD138 positive cells. Rev Bras Hematol Hemoter

2016;38:113-120.

8. Munshi NC, Anderson KC, Bergsagel PL, Shaughnessy J, Palumbo A, Durie B,

Fonseca R, Stewart AK, Harousseau JL, Dimopoulos M, Jagannath S, Hajek R,

Sezer O, Kyle R, Sonneveld P, Cavo M, Rajkumar SV, San Miguel J, Crowley

J, Avet-Loiseau H; International Myeloma Workshop Consensus Panel 2.

Consensus recommendations for risk stratification in multiple myeloma:

report of the International Myeloma Workshop Consensus Panel 2. Blood

2011;117:4696-4700.

9. Morgan G, Davies F, Linet M. Myeloma aetiology and epidemiology. Biomed

Pharmacother 2002;56:223-234.

10. Sailer M, Vykoupil KF, Peest D, Coldewey R, Deicher H, Georgii A. Prognostic

relevance of a histologic classification system applied in bone marrow

biopsies from patients with multiple myeloma: a histopathological

evaluation of biopsies from 153 untreated patients. Eur J Haematol

1995;54:137-146.

11. Kuter DJ, Bain B, Mufti G, Bagg A, Hasserjian RP. Bone marrow fibrosis:

pathophysiology and clinical significance of increased bone marrow stromal

fibres. Br J Haematol 2007;139:351-362.

12. Tefferi A. Pathogenesis of myelofibrosis with myeloid metaplasia. J Clin

Oncol 2005;23:8520-8530.

13. Babarović E, Valković T, Štifter S, Budisavljević I, Seili-Bekafigo I, Duletić-

Načinović A, Lučin K, Jonjić N. Assessment of bone marrow fibrosis and

angiogenesis in monitoring patients with multiple myeloma. Am J Clin

Pathol 2012;137:870-878.

14. Paul B, Zhao Y, Loitsch G, Feinberg D, Mathews P, Barak I, Dupuis M, Li

Z, Rein L, Wang E, Kang Y. The impact of bone marrow fibrosis and JAK2

expression on clinical outcomes in patients with newly diagnosed multiple

myeloma treated with immunomodulatory agents and/or proteasome

inhibitors. Cancer Med 2020;9:5869-5880.

15. Fonseca R, Barlogie B, Bataille R, Bastard C, Bergsagel PL, Chesi M, Davies

FE, Drach J, Greipp PR, Kirsch IR, Kuehl WM, Hernandez JM, Minvielle S,

Pilarski LM, Shaughnessy JD Jr, Stewart AK, Avet-Loiseau H. Genetics

and cytogenetics of multiple myeloma: a workshop report. Cancer Res

2004;64:1546-1558.

16. Smadja NV, Bastard C, Brigaudeau C, Leroux D, Fruchart C. Hypodiploidy is

a major prognostic factor in multiple myeloma. Blood 2001;98:2229-2238.

17. Van Wier S, Braggio E, Baker A, Ahmann G, Levy J, Carpten JD, Fonseca

R. Hypodiploid multiple myeloma is characterized by more aggressive

molecular markers than non-hyperdiploid multiple myeloma. Haematologica

2013;98:1586-1592.

18. Fonseca R, Bergsagel PL, Drach J, Shaughnessy J, Gutierrez N, Stewart AK,

Morgan G, Van Ness B, Chesi M, Minvielle S, Neri A, Barlogie B, Kuehl WM,

Liebisch P, Davies F, Chen-Kiang S, Durie BG, Carrasco R, Sezer O, Reiman

T, Pilarski L, Avet-Loiseau H; International Myeloma Working Group.

International Myeloma Working Group molecular classification of multiple

myeloma: spotlight review. Leukemia 2009;23:2210-2221.

19. Boyd KD, Ross FM, Chiecchio L, Dagrada GP, Konn ZJ, Tapper WJ, Walker

BA, Wardell CP, Gregory WM, Szubert AJ, Bell SE, Child JA, Jackson GH,

Davies FE, Morgan GJ; NCRI Haematology Oncology Studies Group. A novel

prognostic model in myeloma based on co-segregating adverse FISH lesions

and the ISS: analysis of patients treated in the MRC Myeloma IX trial.

Leukemia 2012;26:349-355.

20. Teoh PJ, Chung TH, Sebastian S, Choo SN, Yan J, Ng SB, Fonseca R, Chng WJ.

p53 haploinsufficiency and functional abnormalities in multiple myeloma.

Leukemia 2014;28:2066-2074.

21. Chng WJ, Price-Troska T, Gonzalez-Paz N, Van Wier S, Jacobus S, Blood E,

Henderson K, Oken M, Van Ness B, Greipp P, Rajkumar SV, Fonseca R. Clinical

significance of TP53 mutation in myeloma. Leukemia 2007;21:582-584.

22. Lodé L, Eveillard M, Trichet V, Soussi T, Wuillème S, Richebourg S,

Magrangeas F, Ifrah N, Campion L, Traullé C, Guilhot F, Caillot D, Marit G,

Mathiot C, Facon T, Attal M, Harousseau JL, Moreau P, Minvielle S, Avet-

Loiseau H. Mutations in TP53 are exclusively associated with del(17p) in

multiple myeloma. Haematologica 2010;95:1973-1976.

23. Boyd KD, Ross FM, Tapper WJ, Chiecchio L, Dagrada G, Konn ZJ, Gonzalez D,

Walker BA, Hockley SL, Wardell CP, Gregory WM, Child JA, Jackson GH, Davies

FE, Morgan GJ; NCRI Haematology Oncology Studies Group. The clinical

impact and molecular biology of del(17p) in multiple myeloma treated with

conventional or thalidomide‐based therapy. Genes Chromosomes Cancer

2011;50:765-774.

24. Fonseca R, Blood E, Rue M, Harrington D, Oken MM, Kyle RA, Dewald GW,

Van Ness B, Van Wier SA, Henderson KJ, Bailey RJ, Greipp PR. Clinical and

biologic implications of recurrent genomic aberrations in myeloma. Blood

2003;101:4569-4575.

25. Chiecchio L, Protheroe RK, Ibrahim AH, Cheung KL, Rudduck C, Dagrada GP,

Cabanas ED, Parker T, Nightingale M, Wechalekar A, Orchard KH, Harrison

CJ, Cross NC, Morgan GJ, Ross FM. Deletion of chromosome 13 detected

by conventional cytogenetics is a critical prognostic factor in myeloma.

Leukemia 2006;20:1610-1617.

26. Gutiérrez NC, Castellanos MV, Martín ML, Mateos MV, Hernández JM,

Fernández M, Carrera D, Rosiñol L, Ribera JM, Ojanguren JM, Palomera L,

Gardella S, Escoda L, Hernández-Boluda JC, Bello JL, de la Rubia J, Lahuerta

JJ, San Miguel JF; GEM/PETHEMA Spanish Group. Prognostic and biological

implications of genetic abnormalities in multiple myeloma undergoing

autologous stem cell transplantation: t(4;14) is the most relevant adverse

prognostic factor, whereas RB deletion as a unique abnormality is not

associated with adverse prognosis. Leukemia 2007;21:143-150.

27. Bergsagel PL, Kuehl WM. Chromosome translocations in multiple myeloma.

Oncogene 2001;20:5611-5622.

28. Sonneveld P, Avet-Loiseau H, Lonial S, Usmani S, Siegel D, Anderson KC,

Chng WJ, Moreau P, Attal M, Kyle RA, Caers J, Hillengass J, San Miguel

J, van de Donk NW, Einsele H, Bladé J, Durie BG, Goldschmidt H, Mateos

MV, Palumbo A, Orlowski R. Treatment of multiple myeloma with high-risk

cytogenetics: a consensus of the International Myeloma Working Group.

Blood 2016;127:2955-2962.

29. Avet-Loiseau H, Moreau P, Mathiot C, Charbonnel C, Facon T, Attal M,

Hulin C, Marit G, Minvielle S, Harousseau J; Intergroupe Francophone du

Myélome. Use of bortezomib to overcome the poor prognosis of t(4;14), but

not del(17p), in young patients with newly diagnosed multiple myeloma. J

Clin Oncol 2010;28(15 Suppl):8113.

115


Şeyhanlı A. et al: Genetic and Bone Marrow Features in Multiple Myeloma

Turk J Hematol 2022;39:109-116

30. Shaughnessy JD Jr, Zhan F, Burington BE, Huang Y, Colla S, Hanamura I,

Stewart JP, Kordsmeier B, Randolph C, Williams DR, Xiao Y, Xu H, Epstein

J, Anaissie E, Krishna SG, Cottler-Fox M, Hollmig K, Mohiuddin A, Pineda-

Roman M, Tricot G, van Rhee F, Sawyer J, Alsayed Y, Walker R, Zangari

M, Crowley J, Barlogie B. A validated gene expression model of high-risk

multiple myeloma is defined by deregulated expression of genes mapping

to chromosome 1. Blood 2007;109:2276-2284.

31. Decaux O, Lodé L, Magrangeas F, Charbonnel C, Gouraud W, Jézéquel P,

Attal M, Harousseau JL, Moreau P, Bataille R, Campion L, Avet-Loiseau H,

Minvielle S; Intergroupe Francophone du Myélome. Prediction of survival in

multiple myeloma based on gene expression profiles reveals cell cycle and

chromosomal instability signatures in high-risk patients and hyperdiploid

signatures in low-risk patients: a study of the Intergroupe Francophone du

Myélome. J Clin Oncol 2008;26:4798-4805.

32. Kuiper R, Broyl A, de Knegt Y, van Vliet MH, van Beers EH, van der Holt B,

el Jarari L, Mulligan G, Gregory W, Morgan G, Goldschmidt H, Lokhorst HM,

van Duin M, Sonneveld P. A gene expression signature for high-risk multiple

myeloma. Leukemia 2012;26:2406-2413.

116


RESEARCH ARTICLE

DOI: 10.4274/tjh.galenos.2022.2022.0084

Turk J Hematol 2022;39:117-129

Combination of Haploidentical Hematopoietic Stem Cell

Transplantation with Umbilical Cord-Derived Mesenchymal Stem

Cells in Patients with Severe Aplastic Anemia: A Retrospective

Controlled Study

Şiddetli Aplastik Anemisi Olan Hastaların Göbek Kordonu Kaynaklı Mezenkimal Kök

Hücreleri ile Haplo Uyumlu Hematopoetik Kök Hücre Naklinin Kombinasyonu: Geriye Dönük,

Kontrollü Bir Çalışma

Xian-Fu Sheng*, Hui Li*, Li-Li Hong, Hai-Feng Zhuang

Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China

*These authors contributed equally to this work.

Abstract

Objective: We retrospectively compared the outcomes of patients

with severe aplastic anemia (SAA) who received haploidentical

hematopoietic stem cell transplantation (haplo-HSCT) combined or

not combined with umbilical cord-derived mesenchymal stem cells

(UC-MSCs).

Materials and Methods: A total of 101 patients with SAA were

enrolled in this study and treated with haplo-HSCT plus UC-MSC

infusion (MSC group, n=47) or haplo-HSCT alone (non-MSC group,

n=54).

Results: The median time to neutrophil engraftment in the MSC

and non-MSC group was 11 (range: 8-19) and 12 (range: 8-23) days,

respectively (p=0.049), with a respective cumulative incidence (CI) of

97.82% and 97.96% (p=0.101). Compared to the non-MSC group, the

MSC group had a lower CI of chronic graft-versus-host disease (GVHD)

(8.60±0.25% vs. 24.57±0.48%, p=0.048), but similar rates of grades II-

IV acute GVHD (23.40±0.39% vs. 24.49±0.39%, p=0.849), grades III-IV

acute GVHD (8.51±0.17% vs. 10.20±0.19%, p=0.765), and moderatesevere

chronic GVHD (2.38±0.06% vs. 7.45±0.18%, p=0.352) were

observed. The estimated 5-year overall survival (OS) rates were

78.3±6.1% and 70.1±6.3% (p=0.292) while the estimated 5-year

GVHD-free, failure-free survival (GFFS) rates were 76.6±6.2% and

56.7±6.9% (p=0.045) in the MSC and non-MSC groups, respectively.

Conclusion: In multivariate analysis, graft failure was the only adverse

predictor for OS. Meanwhile, graft failure, grades III-IV acute GVHD,

and moderate-severe chronic GVHD could predict worse GFFS. Our

results indicated that haplo-HSCT combined with UC-MSCs infusion

was an effective and safe option for SAA patients.

Keywords: Severe aplastic anemia, Haploidentical, Hematopoietic

stem cell transplantation, Mesenchymal stem cells

Öz

Amaç: Göbek kordonundan elde edilen mezenkimal kök hücrelerin (UK-

MKH’ler) infüzyonu ile kombine edilmiş ve edilmemiş, haploidentik

hematopoietik kök hücre nakli (haplo-HKHN) yapılan şiddetli aplastik

anemili (ŞAA) hastalarının sonuçları geriye dönük olarak karşılaştırıldı.

Gereç ve Yöntemler: Bu çalışmaya ŞAA’lı toplam 101 hasta alındı

ve hastalar haplo-HSCT ile birlikte UK-MKH infüzyonu (MKH grubu,

N=47) ve tek başına haplo-HKHN (MKH olmayan grup, N=54) şeklinde

iki gruba ayrıldı.

Bulgular: MKH alan ve almayan gruplarda nötrofil engraftmanı için

medyan süreler sırasıyla, 11 (aralık, 8-19) ve 12 (aralık, 8-23) gün

(p=0,049) ve kümülatif insidans (CI) sırası ile %97,82 ve %97,96

(p=0,101) bulundu. MKH almayan grupla karşılaştırıldığında, MKH

alan grupta daha düşük kronik graft-versus-host hastalığı (cGVHD)

CI saptandı (%8,60±0,25’e karşı %24,57±0,48, p=0,048), ancak II-

IV akut GVHD (aGVHD) (%23,40±0,39-%24,49±0,39, p=0,849), III-

IV aGVHD (%8,51±0,17 ve %10,20±0,19, p=0,765) ve orta-şiddetli

cGVHD (%2,38±0,06 vs. %7,45±0,18, p=0,352) gruplarında fark

görülmedi. MKH alan ve MKH almayan gruplarda tahmini beş yıllık

genel sağkalım oranları %78,3±6,1 ve %70,1±6,3 (p=0,292) iken,

GVHD’siz, hastalıksız sağkalım (GFFS) oranları sırasıyla, %76,6 ±%6,2

ve %56,7±6,9 (p=0,045) idi.

Sonuç: Çok değişkenli analizde greft yetmezliği, OS için tek olumsuz

gösterge idi. Bu arada, greft yetmezliği, III-IV aGVHD ve orta-şiddetli

cGVHD arasında daha kötü GFFS’yi öngörebilir. Sonuçlarımız, UC-MKH

infüzyonu ile birlikte haplo-HKHN’nin ŞAA hastaları için etkili ve

güvenli bir seçenek olduğunu gösterdi.

Anahtar Sözcükler: Şiddetli aplastik anemi, Haplo uyumlu,

Hematopoetik kök hücre nakli, Mezenkimal kök hücre

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Hai-Feng Zhuang, M.D., Zhejiang Chinese Medical University,

Hangzhou, Zhejiang, China

Phone : +86 13858159652

E-mail : zhuanghaifeng5@163.com ORCID: orcid.org/0000-0003-2802-7910

Received/Geliş tarihi: March 2, 2022

Accepted/Kabul tarihi: April 21, 2022

117


Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

Turk J Hematol 2022;39:117-129

Introduction

Severe aplastic anemia (SAA) is a rare, life-threatening bone

marrow (BM) failure syndrome characterized by pancytopenia

and hypoplastic bone marrow. The incidence is 2-2.4 per million

per year in Europe but higher in China, affecting 7.4 patients

per million [1].

Although hematopoietic stem cell transplantation (HSCT) from

a human leukocyte antigen (HLA)-matched related donor (MRD)

is the first choice for SAA patients who are <40 years old, it

is difficult to search for HLA-matched siblings in China [2].

Immunosuppressive therapy (IST) with antithymocyte globulin

(ATG) and cyclosporin A (CsA) is recommended as the first-line

treatment for SAA patients who lack an appropriate MRD and

those aged >40 years [3]. However, most patients cannot be

cured by IST. Previous studies showed that response rates were

in the range of 40%-60% and 25% of responders subsequently

relapsed [4,5,6]. Therefore, the treatment of SAA patients

who lack sibling donors or fail to respond to IST is extremely

challenging.

Stem cells from haploidentical family donors have become the

most common source of HSCT in China due to the advantage of

immediate availability for almost any patient [7]. Haploidentical

HSCT (haplo-HSCT) has greatly developed in recent years. With

improvements in donor selection, conditioning regimens, and

graft-versus-host disease (GVHD) prophylaxis, studies comparing

haplo-HSCT with MRD-HSCT in young patients have revealed

limited differences in overall survival (OS) [6,8,9]. However, the

higher incidence of graft failure (GF) and GVHD has limited the

clinical application of haplo-HSCT for SAA patients [6,10,11,12].

Mesenchymal stem cells (MSCs) have been shown to support

hematopoiesis and display potent immunomodulatory

properties for treating GVHD after HSCT [13,14]. It was reported

that human umbilical cord-derived MSCs (UC-MSCs) have

higher activity levels of proliferation and differentiation in

comparison with bone marrow-derived MSCs (BM-MSCs) [11].

Thus, UC-MSCs are highly promising for use in haplo-HSCT. In

recent years, some studies have reported the co-transplantation

of haplo-HSCT and UC-MSCs with favorable outcomes in cases

of SAA [11,12,15,16]. To the best of our knowledge, there are

still no head-to-head studies exploring the efficiency and

safety of haplo-HSCT with and without UC-MSCs. In the present

study, we report the outcomes of haplo-HSCT with or without

third-party UC-MSCs for the treatment of SAA patients in our

centers from June 2014 to June 2021.

Materials and Methods

Patients

Between June 2014 and June 2021, 101 consecutive SAA

patients who underwent haplo-HSCT in our transplant unit

were enrolled in this study. Informed consent was obtained from

all patients or their parents and the study was approved by the

Ethics Committee of the First Affiliated Hospital of Zhejiang

Chinese Medical University. All patients met the following

inclusion criteria: (1) diagnosed with SAA or very severe aplastic

anemiaSAA according to the guidelines of the International

Aplastic Anemia Study Group; (2) no HLA-identical sibling

donor; (3) no serious infectious disease or acute hemorrhaging;

(4) absence of severe liver, renal, lung, and heart diseases;

(5) Eastern Cooperative Oncology Group score of 0-2 points.

HLA compatibility was determined by high-resolution DNA

techniques for the HLA-A, B, C, DRB1, and DQB1 loci. Donors

were ranked on the basis of HLA match, age (younger preferred),

gender (same preferred), and health status (better preferred).

Conditioning Regimen

Patients who underwent haplo-HSCT were placed on regimens

including fludarabine (Flu)/cyclophosphamide (Cy)/ATG or

Bu/Cy/ATG. Patients with acute SAA received the following

regimen: intravenous fFlu at 30 mg/m 2 /day on days -5 and

-2; intravenous c (Cy) at 50 mg/kg/day on days -5 to -2; and

intravenous ATG (rabbit, Genzyme Polyclonals SAS, Lyon, France)

at 2.5 mg/kg/day on days -5 to -2. For patients with chronic

SAA (SAA-II), or, in other words, patients who had progressed to

SAA from non-SAA [17], the same protocol for Cy and ATG was

applied as above with the addition of busulfan (Bu) at 3.2 mg/

kg/day from day -7 to -6.

All patients underwent testing for donor-specific anti-HLA

antibodies (DSA) before transplantation. If the DSA results were

positive (MFI >5000), the patient received rituximab (once a

week, 4 times) and plasmapheresis (2-3 times). The DSA results

were then reviewed again and transplantation occurred if the

value dropped below 5000. Among the 101 patients enrolled in

this study, 5 patients had positive DSA results. After treatment,

all of these patients had negative DSA results.

Stem Cell Collection and Infusion

All donors were injected with granulocyte colony-stimulating

factor (G-CSF) subcutaneously at 10 µg/kg/day for 4-5

continuous days. BM grafts were collected by BM aspiration in

an operating room on day +1. The target volume was 10 to 20

mL/kg of the recipient’s weight. In cases of ABO incompatibility,

red blood cells were removed by sedimentation with Hespan.

Peripheral blood stem cells (PBSCs) were collected with a COBE

Spectra device (Gambro BCT, Lakewood, CO, USA) on day +2. The

target mononuclear cell) count from BM and peripheral blood

was ≥5x10 8 /kg and the target CD34 + cell count was ≥2x10 6 /kg

of the recipient’s weight. An additional collection of PBSCs was

performed on day +3 if the initial cell numbers were insufficient.

UC-MSCs were purchased from the Shanghai Cord Blood Bank.

A total of 1x10 6 /kg UC-MSCs was infused 4 h before the infusion

of the graft on day +1.

118


Turk J Hematol 2022;39:117-129

Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

GVHD Prophylaxis

In the protocol applied for these patients, prophylaxis for acute

GVHD (aGVHD) included CsA, mycophenolate mofetil (MMF),

and short-term methotrexate (MTX). CsA was administrated

intravenously twice daily at a dose of 1.5 mg/kg/day from day

-7 until bowel function returned to normal, at which time

patients received oral CsA. A target trough blood concentration

of 200-300 ng/mL was maintained for ≥1 year after

haplo-HSCT. CsA was gradually tapered thereafter and was

withdrawn completely in the following 2-3 months. MMF was

given orally at 500 mg every 12 h (250 mg for children) from

day -7 to +30 and subsequently at 250 mg from day +30 to

+90. MTX was administrated intravenously at a dose of 15

mg/m2 on day +1 and 10 mg/m2 on days +3, +6, and +11. If

GVHD occurred in any organ, CsA and MMF were continued and

adjusted to therapeutic concentrations.

Definitions and Post-transplantation Evaluations

Neutrophil engraftment was defined as occurring on the first

of 3 consecutive days with an absolute neutrophil count of

>0.5x10 9 /L. Platelet engraftment was defined as occurring on the

first day of a platelet count of >20x10 9 /L without transfusion

for 7 consecutive days. Short-tandem repeat polymerase chain

reaction of peripheral whole blood was performed monthly to

assess hematopoietic chimerism from the time of neutrophil

recovery.

Primary GF (PGF) was defined as the failure to achieve

neutrophil engraftment before day +28 after transplantation.

Secondary GF was defined as graft loss after initial engraftment,

with the recipient experiencing pancytopenia and hypocellular

BM without moderate to severe aGVHD [15]. Delayed platelet

recovery was defined as platelet engraftment achieved after day

+28. aGVHD was defined according to the criteria proposed by

the 1994 Consensus Conference on aGVHD Grading [18], while

chronic GVHD (cGVHD) was defined according to the National

Institutes of Health Consensus Conference on cGVHD [19].

Diagnoses of cytomegalovirus (CMV) infection, CMV pneumonia,

Epstein-Barr virus (EBV) infection, and EBV-associated posttransplant

lymphoproliferative disorders (PTLDs) were based on

standard clinical criteria.

OS was defined as the time from the date of haplo-HSCT to the

date of death or last follow-up. GVHD-free, failure-free survival

(GFFS) was defined as survival without grades III-IV aGVHD,

moderate-severe cGVHD, or HSCT failure. Death, primary or

secondary GF, and relapse were considered as HSCT failures.

Supportive Care

All patients were admitted to class 100 laminar flow clean

wards after a skin-care bath. Patients received oral antibiotics

(levofloxacin for adults, gentamycin for children) for

gastrointestinal decontamination before transplantation.

Fluconazole, acyclovir, and trimethoprim/sulfamethoxazole

were administered to prevent fungal, viral, and Pneumocystis

pneumonia infection, respectively. Heparin and prostaglandin E1

were administrated to prevent veno-occlusive disease. Human

immunoglobulin was administered intravenously at a dose of 10

g once a week from day 0 to day +100 after the transplantation.

Recombinant human G-CSF was given at a dose of 5 µg/kg/day

from day +3 until neutrophil recovery. Irradiated erythrocytes

and platelets were given to maintain a hemoglobin level of >60

g/L and a platelet count of >20x10 9 /L.

Statistical Analysis

The date of last follow-up for all surviving patients was December

31, 2021. Patient baseline characteristics were compared by

chi-square test for categorical variables and by Mann-Whitney U

and Kruskal-Wallis tests for continuous data. OS and GFFS were

calculated using the Kaplan-Meier method and the groups were

compared using the log-rank test. The cumulative incidence

(CI) method was used to calculate the incidence of aGVHD and

cGVHD with death and GF as competing risks according to the

competing risk model. We used univariate and multivariate

analyses to determine whether any of the selected factors were

predictive of OS and GFFS. In multivariate analysis, all factors

with values of p<0.1 in univariate analysis were included in the

Cox regression model. Values of p<0.05 were considered to be

significant. SPSS 19.0 (IBM Corp., Armonk, NY, USA) was used

for statistical analyses.

Results

Characteristics of Patients and Donors

SAA patients undergoing haplo-HSCT with UC-MSC infusion

(MSC group, n=47) or without UC-MSC infusion (non-MSC

group, n=54) were enrolled in this study. Characteristics of the

patients and their donors were compared between groups as

shown in Table 1. The two cohorts were similar with regard to

age distribution, sex ratio, disease severity, interval between

diagnosis and transplantation, and previous treatments. No

differences were found in the baseline characteristics of donors

and grafts between the two groups.

Engraftment

Forty-seven patients (100%) in the MSC group and 49 patients

(90.74%) in the non-MSC group survived for 28 days. The five

patients who experienced early mortality before engraftment

were not considered in the subsequent analysis. One patient

in each group experienced PGF. Two patients died suddenly

before a second HSCT could be performed. The CIs of 28-day

neutrophil engraftment were 97.82±0.06% and 97.96±0.06%

in the MSC and non-MSC groups, respectively (p=0.101).

119


Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

Turk J Hematol 2022;39:117-129

Table 1. Characteristics of patients with severe anaplastic anemia, their donors, and grafts.

Variable MSC group (n=47) Non-MSC group (n=54) p

Median age, years (range)

≤20 years, n (%)

21-39 years, n (%)

≥40 years, n (%)

Sex, n (%)

Male

Female

Disease severity, n (%)

SAA

VSAA

SAA-PNH

25 (8-56)

15 (31.9)

26 (55.3)

6 (12.8)

24 (51.1)

23 (48.9)

23(48.9)

21 (44.7)

3 (6.4)

33 (9-55)

14 (25.9)

24 (44.4)

22 (21.8)

33 (61.1)

21 (38.9)

35 (64.8)

17 (31.5)

2 (3.7)

Median time from diagnosis to HSCT, months (range) 3 (1-300) 7 (1-120) 0.502

Previous treatment, n (%)

CsA ± Andriol

ATG/ALG ± CsA ± Andriol

Supportive care

34 (72.3)

7 (14.9)

6 (12.8)

43 (79.6)

2 (3.7)

9 (16.7)

0.145

Donor-recipient sex match, n (%)

Male-male

Male-female

Female-male

Female-female

15 (31.9)

11 (23.4)

9 (19.1)

12 (25.5)

18 (33.3)

7 (13.0)

15 (27.8)

14 (25.9)

Donor-recipient relationship, n (%) 0.574

Mother-child 5 (10.6) 5 (9.3)

Father-child 12 (25.5) 10 (18.5)

Child-mother 6 (12.8) 10 (18.5)

Child-father

Siblings

5 (10.6)

19 (40.4)

11 (20.4)

18 (33.3)

HLA mismatched, n (%) 0.950

Haplo 5 locus mismatched

Haplo 4 locus mismatched

Haplo 3 locus mismatched

Haplo 2 locus mismatched

Haplo 1 locus mismatched

ABO blood types match, n (%)

Matched

Major unmatched

Minor unmatched

Major and minor unmatched

Conditioning regimen, n (%)

Flu/Cy/ATG

Bu/Cy/ATG

Graft type, n (%)

BM + PB

PB

Stem cells

MNCs, x10 8 /kg, median (range)

CD34 + , x10 6 /kg, median (range)

30 (63.8)

8 (17.0)

7 (14.9)

1 (2.1)

1 (2.1)

27 (57.4)

6 (12.8)

10 (21.3)

4 (8.5)

35 (74.5)

12 (25.5)

47 (100.0)

0

8.68 (2.49-21.80)

4.90 (1.92-12.70)

31 (57.4)

12 (22.2)

9 (16.7)

1 (1.9)

1 (1.9)

33 (61.1)

9 (16.7)

8 (14.8)

4 (7.4)

45 (83.3)

9 (16.7)

52 (96.3)

2 (3.7)

7.33 (2.35-26.00)

5.21 (1.98-16.86)

MSCs: Mesenchymal stem cells; SAA: severe aplastic anemia; VSAA: very severe aplastic anemia; PNH: paroxysmal nocturnal hemoglobinuria; HSCT: hematopoietic stem cell

transplantation; CsA: cyclosporin A; ATG: antithymocyte globulin; ALG: antilymphocyte globulin; HLA: human leukocyte antigen; Haplo: haploidentical; Flu: fludarabine; Cy:

cyclophosphamide; Bu: busulfan; BM: bone marrow; PB: peripheral blood; MNCs: mononuclear cells. Values of p<0.05 were considered to be significant.

0.134

0.123

0.310

0.256

0.505

0.815

0.273

0.497

0.251

0.817

120


Turk J Hematol 2022;39:117-129

Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

The median times to neutrophil engraftment were 11

(range: 8-19) and 12 (range: 8-23) days, respectively (p=0.049).

Forty-three and 48 patients achieved platelet engraftment in

the MSC and non-MSC groups, respectively, with respective

CIs of platelet engraftment of 91.49±0.19% and 91.84±0.16%

(p=0.345). The median times to platelet engraftment were 12

(range: 7-43) and 14 (range: 6-70) days, respectively (p=0.047).

In the MSC group, 2 patients (4.26%) experienced secondary

GF and 3 patients (6.38%) had delayed platelet recovery. In the

non-MSC group, 3 (6.12%) and 6 (12.24%) patients experienced

secondary GF and delayed platelet recovery, respectively. The

patients who experienced secondary GF subsequently died due

to infection or cerebral hemorrhage. No significant differences

were observed between groups in terms of PGF, secondary GF, or

delayed platelet recovery (p>0.05, Table 2).

GVHD

Table 2 presents the incidence and severity of GVHD for both

groups. Patients with primary engraftment were included in

the aGVHD analysis. In the MSC group, 17 patients experienced

aGVHD after HSCT. These included 6 patients with grade I, 7

with grade II, 3 with grade III, and 1 with grade IV aGVHD. Of

the 49 patients in the non-MSC groups, 21 experienced aGVHD

after HSCT, including 9 with grade I, 7 with grade II, 4 with

Table 2. Comparison of clinical outcomes between the groups after haploidentical hematopoietic stem cell transplantation.

Variable MSC group (n=47) Non-MSC group (n=54) p

Patients surviving longer than 30 days, n (%) 47 (100) 49 (90.7) 0.059

Engraftment

Median myeloid recovery, days (range) 11 (8-19) 12 (8-23) 0.049

Median platelet recovery, days (range) 12 (7-43) 14 (6-70) 0.047

Primary graft failure, n (%) 1 (2.1) 1 (2.0) 1.000

Secondary graft failure, n (%) 2 (4.3) 3 (6.1) 1.000

Delayed platelet recovery, n (%) 3 (6.4) 6 (12.2) 0.526

Acute GVHD n=47 n=49

None, n (%) 30 (63.8) 28 (57.1) 0.503

II-IV, n (%) 11 (23.4) 12 (24.5) 0.901

III-IV, n (%) 4 (8.7) 5 (10.2) 1.000

Patients surviving longer than 100 days, n (%) 42 (89.4) 44 (81.5) 0.267

Chronic GVHD n=42 n=44

None, n (%) 38 (90.5) 34 (77.3) 0.097

Mild, n (%) 2 (4.8) 7 (15.9) 0.182

Moderate to severe, n (%) 2 (4.8) 3 (6.8) 1.000

Infection, n (%) n=47 n=54

Viremia

CMV 14 (29.8) 15 (27.8) 0.824

EBV 31 (66.0) 30 (55.6) 0.286

EBV-associated PTLD 3 (6.4) 4 (7.4) 1.000

Pulmonary infections 14 (29.8) 16 (29.6) 0.986

Septicemia 7 (14.9) 11 (20.4) 0.473

Herpes zoster 2 (4.3) 10 (18.5) 0.027

Causes of death, n (%) n=47 n=54

Infection 4 (8.5) 5 (9.3) 1.000

RRTs 0 3 (5.6) 0.246

GF 2 (4.3) 3 (5.6) 1.000

GVHD 3 (6.4) 2 (3.7) 0.873

TMA 1 (2.1) 0 0.465

Leukoencephalopathy syndrome 0 1 (1.9) 1.000

Median follow-up time among living patients, months (range) 30.8 (6.5-91.0) 28.1 (6.8-84.5) 0.427

MSCs: Mesenchymal stem cells; GVHD: graft-versus-host disease; CMV: cytomegalovirus; EBV: Epstein-Barr virus; PTLD: post-transplant lymphoproliferative disorder; RRTs: regimenrelated

toxicities; GF: graft failure; TMA: thrombotic microangiopathy. Values of p<0.05 were considered to be significant.

121


Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

Turk J Hematol 2022;39:117-129

grade III, and 1 with grade IV. At 100 days after transplantation,

the CIs of grade II-IV aGVHD for the MSC and non-MSC groups

were 23.40±0.39% and 24.49±0.39%, respectively (p=0.849,

Figure 1a). The CIs of grade III-IV aGVHD in the MSC and

non-MSC groups were 8.51±0.17% and 10.20±0.19%,

respectively (p=0.765, Figure 1b).

Forty-two patients in the MSC group and 44 in the non-MSC

group with survival longer than 100 days after transplantation

were assessed for cGVHD. In the MSC group, 2 patients had mild

cGVHD, 2 had moderate cGVHD, and none had severe cGVHD. In

the non-MSC group, 7 patients had mild cGVHD, 2 had moderate

cGVHD, and 1 had severe cGVHD. Patients in the MSC group

had a lower CI of cGVHD than the non-MSC group (8.60±0.25%

vs. 24.57±0.48%, p=0.048, Figure 1c). However, there was no

significant difference in the CIs of moderate-severe cGVHD

between the two groups (2.38±0.06% vs. 7.45±0.18%, p=0.352,

Figure 1d).

Infectious Complications

The most common infections after transplantation were the

reactivation of CMV and EBV, pulmonary infections, septicemia,

and herpes zoster (Table 2).

Fourteen of the 47 patients (29.8%) in the MSC group

experienced CMV reactivation following transplantation as

detected by DNA testing. In the non-MSC group, 15 of 54

patients (27.8%) experienced CMV reactivation. There was no

Figure 1. Cumulative incidences of graft-versus-host disease (GVHD) for the two groups. a) Cumulative incidences of grade II-IV acute

GVHD (aGVHD): mesenchymal stem cell (MSC) group 23.40±0.39% vs. non-MSC group 24.49±0.39% (p=0.849). b) Cumulative incidences

of grade III-IV aGVHD: MSC group 8.51±0.17% vs. non-MSC group 10.20±0.19% (p=0.765). c) Cumulative incidences of chronic GVHD

(cGVHD): MSC group 8.60±0.25% vs. non-MSC group 24.57±0.48% (p=0.048). d) Cumulative incidences of moderate-severe cGVHD:

MSC group 2.38±0.06% vs. non-MSC group 7.45±0.18% (p=0.352).

HSCT: Hematopoietic stem cell transplantation.

122


Turk J Hematol 2022;39:117-129

Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

significant difference in CMV reactivation between the two

groups (p=0.824, Table 2). Viral infections were treated with

foscarnet or ganciclovir and gamma globulin. In the non-MSC

group, one patient developed CMV retinitis and recovered with

an infusion of CMV-specific cytotoxic T lymphocytes. Other

patients who did not develop CMV disease were mostly cured

after antiviral treatment.

As shown in Table 2, 31 patients in the MSC group (66.0%)

and 30 patients in the non-MSC group (55.6%) experienced

EBV reactivation (p=0.286). Nineteen patients (10 in the MSC

group and 9 in the non-MSC group) who were not successfully

treated with antiviral drugs were treated with rituximab. EBV

copy numbers decreased to the normal range in most cases.

However, one patient in the non-MSC group only recovered with

EBV-specific cytotoxic T lymphocytes. Three patients (6.4%)

and 4 patients (7.4%) progressed to EBV-associated PTLD,

respectively, in the MSC and non-MSC groups (p=1.000). All

patients with PTLD received rituximab treatment, and some

patients received it in combination with chemotherapy. Three

patients with PTLD in the non-MSC group died of complications

resulting from immunochemotherapy on day +274, day +205,

and day +134, respectively. Others recovered from PTLD with the

regression of enlarged lymph nodes and EBV copy numbers that

declined to normal values.

In the MSC group, 14 patients (29.8%) experienced pulmonary

infections, 7 patients (14.9%) experienced septicemia, and

2 patients (4.3%) experienced herpes zoster. In the non-MSC

group, 16 patients (29.6%) experienced pulmonary infections,

11 patients (20.4%) experienced septicemia, and 10 patients

(18.5%) experienced herpes zoster. The incidence of herpes

zoster was higher in the non-MSC group than the MSC group

(p=0.027), but there was no significant difference in the

incidence of pneumonia or sepsis between the groups (p>0.05,

Table 2).

3 cases, GVHD in 2 cases (1 each of severe aGVHD and cGVHD),

and leukoencephalopathy syndrome in 1 case.

Survival

The estimated 5-year OS rates in the MSC and non-MSC groups

were 78.3±6.1% and 70.1±6.3%, respectively, and the difference

was not significant (p=0.292, Figure 3a). However, the estimated

5-year GFFS rate in the MSC group was significantly higher

than that of the non-MSC group (76.6±6.2% vs. 56.7±6.9%,

respectively, p=0.045) (Figure 3b). In univariate analysis, age,

disease severity, donor-recipient relationship, graft type, and GF

significantly predicted OS and GFFS (Tables 3 and 4). Meanwhile,

grade II-IV aGVHD, grade III-IV aGVHD, and moderate-severe

cGVHD also predicted worse GFFS (Table 4). In multivariate

Cox regression analysis, only GF was found to be predictive

of worse OS (p=0.000, Table 3), while GF, grade III-IV aGVHD,

and moderate-severe cGVHD were significantly associated with

lower GFFS (p<0.05, Table 4).

Discussion

Haplo-HSCT is usually considered as a salvage treatment option

for SAA patients after failed IST. In recent years, some studies

have evaluated haplo-HSCT as an upfront therapy for newly

diagnosed SAA patients [6,20,21]. The currently available data

reflect a favorable survival outcome of haplo-HSCT in China.

However, higher incidences of GF and GVHD have limited its

clinical applications [7]. Numerous trials have been undertaken

to overcome these challenges, including the co-transplantation

of MSCs [13,22,23,24]. MSCs possess the capacity to differentiate

into various types of cells, modulate immune response, and

Transplantation-Related Mortality

During a median follow-up period of 29.5 (range: 6.5-91)

months, 10 and 14 patients died of transplantation-related

mortality (TRM) in the MSC and non-MSC groups, respectively,

with a median time to death of 91 (range: 31-270) and 76

(range: 1-274) days (p=0.334). The CIs of TRM in the MSC and

non-MSC groups were 21.7±6.1% and 27.2±6.2% (p=0.424,

Figure 2). Severe infection was the primary cause of death in

both groups (Table 2). In the MSC group, 4 patients died of

infection (3 pulmonary infections and 1 case of septicemia), 3

died of GVHD (1 severe aGVHD and 2 severe cGVHD), 2 of GF

(1 PGF and 1 secondary GF), and 1 of thrombotic microangiopathy.

In the non-MSC group, the causes of TRM included infection in

5 cases (4 pulmonary infections and 1 case of septicemia), GF in

3 cases (1 PGF and 2 secondary GF), regimen-related toxicities in

Figure 2. Cumulative incidences of transplantation-related

mortality (TRM) during follow-up in the mesenchymal stem

cell (MSC) group and non-MSC group were 21.7±6.1% and

27.2±6.2%, respectively (p=0.424).

HSCT: Hematopoietic stem cell transplantation.

123


Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

Turk J Hematol 2022;39:117-129

Figure 3. Overall survival and graft-versus-host disease (GVHD)-free, failure-free survival of the two groups. a) Comparison of estimated

5-year overall survival between mesenchymal stem cell (MSC) group and non-MSC group (78.3±6.1% vs. 70.1±6.3%, p=0.292). b)

Comparison of estimated 5-year GVHD-free, failure-free survival between MSC group and non-MSC group (76.6±6.2% vs. 56.7±6.9%,

p=0.045).

HSCT: Hematopoietic stem cell transplantation.

support hematopoiesis. They are currently widely used in cases

of hematological disease, and especially in treating aplastic

anemia and GF, promoting HSC engraftment, and preventing

and suppressing GVHD [25]. Given the encouraging results,

some researchers have tried co-transplanting MSCs with HSCs

in haplo-HSCT for SAA patients [9,12,13,14,26,27]. Due to the

rarity of SAA, however, there are still no trials directly evaluating

the role of MSCs in haplo-HSCT for SAA patients. To the best

of our knowledge, we present the first prospective, head-tohead

study comparing the efficacy and safety of haplo-HSCT

combined with or not combined with MSC infusion. Our results

have demonstrated that co-transplantation of MSC could

shorten the engraftment time, reduce the incidence of cGVHD,

and extended the GFFS.

GF is always a major risk of HSCT in cases of SAA, occurring

more frequently in these cases than in other malignant diseases.

In earlier years, the GF rate was as high as 70% [28]. In recent

years, the GF rate has significantly fallen to less than 10% due

to improved conditioning regimens, the use of G-CSF-mobilized

grafts, and cell therapy strategies such as cord-blood units

and MSC infusions [27,29,30,31,32]. MSCs have been shown to

support hematopoiesis in vitro and in vivo [25]. Some researchers

combined haplo-HSCT with MSC infusions in a series of singlearm

trials, and the promising results included engraftment rates

ranging from 93.2% to 98.9% [9,11,12,13,14,15]. Although the

current incidence of engraftment appears to be higher than

the historically reported values, there are no head-to-head

studies to date to confirm this. Our results showed no statistical

differences in the CIs of neutrophil engraftment (97.82±0.06%

vs. 97.96±0.06%, p=0.101) or platelet engraftment

(91.49±0.19% vs. 91.84±0.16%, p=0.345) in the MSC and non-

124

MSC groups. However, patients in the MSC group experienced

faster neutrophil engraftment (11 (range: 8-19) vs. 12 (range:

8-23) days, p=0.049) and platelet engraftment (12 (range: 7-43)

vs. 14 (range: 6-70) days, p=0.047) compared to those in the

non-MSC group. Our results thus indicate that co-transplantation

of MSCs could enhance engraftment in HSCT.

GVHD is another major challenge in haplo-HSCT for SAA. MSC

applications in the field of GVHD treatment have achieved great

success due to the immunoregulatory properties of these cells

[33,34]. However, the efficacy of MSCs for GVHD prophylaxis

varies in different reports [25,35]. In a phase II study, 37 patients

with hematological malignant diseases were randomly divided

into a standard GVHD prophylaxis group (Group 1) and a group

receiving standard GVHD prophylaxis combined with MSCs

(Group 2). Results showed that the incidence of grade II-IV

aGVHD was lower in Group 2 compared to Group 1 (5.3% vs.

38.9%, p=0.002) [36]. There have been no controlled studies

to date to confirm the efficacy of MSCs for GVHD prophylaxis

in SAA patients, although some studies reported that cotransplantation

of haplo-HSCs and MSCs in SAA patients was

safe and effective [12,13,14,15]. The reported CIs of grade II-IV

aGVHD, grade III-IV aGVHD, and cGVHD were previously found

to be approximately 23.5%-29.3%, 4.9%-11.4%, and 18.2%-

26.8%, respectively [12,13,14,15]. These results did not appear

to differ significantly from the historical findings. On the other

hand, a meta-analysis concluded that MSCs may make little

or no difference for the risk of GVHD compared to treatment

without MSCs in haplo-HSCT for SAA patients [16]. Our results

showed that the CI of cGVHD in the MSC group was significantly

lower compared to that of the non-MSC group (8.60±0.25%

vs. 24.57±0.48%, p=0.048). However, there were no statistical


Turk J Hematol 2022;39:117-129

Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

Table 3. Univariate and multivariate analysis of factors associated with overall survival.

Variable

Age, years

Univariate

OS

Multivariate

HR (95% CI) p HR (95% CI) p

21-39 vs. ≤20 0.544 (0.235-1.285) 0.011 0.296 (0.052-1.671) 0.168

≥40 vs. ≤20 0.185 (0.050-0.684) 0.155 0.674 (0.216-2.106) 0.497

Gender

Male vs. female 1.331 (0.616-2.873) 0.467

Disease severity

VSAA vs. SAA 0.275 (0.078-0.966) 0.044 0.443 (0.100-1.958) 0.283

SAA-PNH vs. SAA 0.346 (0.095-1.258) 0.107 0.418 (0.103-1.691) 0.221

Time from diagnosis to HSCT

≤2 vs. > 2 months 0.767 (0.355-1.654) 0.499

Previous treatment

ATG/ALG vs. CsA 0.843 (0.287-2.479) 0.757

Supportive care vs. CsA 1.280 (0.286-5.719) 0.747

Donor-recipient relationship

Mother-child vs. siblings 0.269 (0.082-0.883) 0.030 0.580 (0.157-2.149) 0.415

Father-child vs. siblings 0.684 (0.171-2.735) 0.591 2.246 (0.448-11.262) 0.325

Child-mother vs. siblings 0.381 (0.107-1.350) 0.135 0.933 (0.214-4.064) 0.927

Child-father vs. siblings 1.186 (0.411-3.420) 0.752 1.042 (0.314-3.455) 0.946

Graft type

PB vs. BM + PB 0.168 (0.040-0.715) 0.016 0.165 (0.025-1.094) 0.062

Treatment group

MSC vs. non-MSC 1.524 (0.691-3.359) 0.296

GF

Yes vs. no 0.073 (0.027-0.197) 0.000 0.080 (0.027-0.237) 0.000

Grade II-IV aGVHD

Yes vs. no 0.831 (0.349-1.978) 0.676

Grade III-IV aGVHD

Yes vs. no 0.491 (0.169-1.426) 0.191

Moderate-severe cGVHD

Yes vs. no 0.712 (0.168-3.014) 0.645

SAA: Severe aplastic anemia; VSAA: very severe aplastic anemia; PNH: paroxysmal nocturnal hemoglobinuria; HSCT: hematopoietic stem cell transplantation; ATG: antithymocyte

globulin; ALG: antilymphocyte globulin; CsA: cyclosporin A; BM: bone marrow; PB: peripheral blood; MSC: mesenchymal stem cell; aGVHD: acute graft-versus-host disease; cGVHD:

chronic GVHD; OS: overall survival; HR: hazard ratio; CI: confidence interval. Values of p<0.05 were considered to be significant.

differences in the CIs of grade II-IV aGVHD, grade III-IV aGVHD,

or moderate-severe cGVHD between the two groups. Another

phase II multicenter, randomized, double-blind controlled

study similarly demonstrated that prophylactic infusion of

MSCs after haplo-HSCT may reduce the incidence of cGVHD

in hematological malignant diseases (27.4% vs. 49%, p=0.021)

[37]. Our data and previous studies thus suggest that MSCs cotransplanted

during HSCT decrease the incidence of cGVHD to

some extent.

Infection is another major obstacle to survival, and particularly

lethal viral infections. Accordingly, another issue of concern

is whether MSCs increase the incidence of infection. In the

past, some studies suggested that MSCs did increase the risk

of infection by suppressing T-cell response and increasing the

secretion of some proinflammatory cytokines [38,39]. However,

our results showed that the incidences of all types of infections

did not increase after MSC administration. The incidence of

CMV and EBV reactivation in the MSC group was 29.8% and

66.6%, respectively, and these values were consistent with some

recent studies [14,40]. However, other studies reported a higher

risk of CMV reactivation (51.7% to 65.9%) and a lower risk

of EBV reactivation (22.7% to 31.8%) [2,12,13], which seems

to be contrary to our results. This may be related to different

125


Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

Turk J Hematol 2022;39:117-129

Table 4. Univariate and multivariate analysis of factors associated with graft-versus-host disease-free, failure-free survival.

Variable

Age, years

Univariate

GFFS

Multivariate

HR (95% CI) p HR (95% CI) p

21-39 vs. ≤20 0.152 (0.049-0.467) 0.001 0.634 (0.133-3.026) 0.567

≥40 vs. ≤20 0.387 (0.187-0.799) 0.010 1.033 (0.363-2.936) 0.951

Gender

Male vs. female 1.291 (0.659-2.530) 0.457

Disease severity

VSAA vs. SAA 0.351 (0.102-1.208) 0.097 0.461 (0.099-2.137) 0.322

SAA-PNH vs. SAA 0.577 (0.167-1.994) 0.384 0.500 (0.116-2.164) 0.354

Time from diagnosis to HSCT

≤2 vs. >2 months 0.633 (0.323-1.241) 0.183

Previous treatment

ATG/ALG vs. CsA 0.866 (0.331-2.263) 0.769

Supportive care vs. CsA 1.331 (0.357-4.958) 0.670

Donor-recipient relationship

Mother-child vs. siblings 0.228 (0.085-0.612) 0.003 0.403 (0.123-1.320) 0.133

Father-child vs. siblings 0.537 (0.165-1.745) 0.301 0.594 (0.120-2.941) 0.523

Child-mother vs. siblings 0.220 (0.068-0.716) 0.012 0.649 (0.149-2.830) 0.565

Child-father vs. siblings 0.949 (0.385-2.335) 0.909 1.495 (0.487-4.583) 0.482

Graft type

PB vs. BM + PB 0.222 (0.053-0.929) 0.039 0.504 (0.086-2.940) 0.446

Treatment group

MSC vs. non-MSC 2.053 (1.001-4.212) 0.050 1.694 (0.731-3.925) 0.219

GF

Yes vs. no 0.125 (0.051-0.311) 0.000 0.049 (0.015-0.162) 0.000

Grade II-IV aGVHD

Yes vs. no 0.447 (0.221-0.906) 0.025 1.022 (0.254-4.110) 0.975

Grade III-IV aGVHD

Yes vs. no 0.064 (0.025-0.161) 0.000 0.023 (0.004-0.137) 0.000

Moderate-severe cGVHD

Yes vs. no 0.242 (0.093-0.629) 0.004 0.128 (0.039-0.414) 0.001

SAA: Severe aplastic anemia; VSAA: very severe aplastic anemia; PNH: paroxysmal nocturnal hemoglobinuria; HSCT: hematopoietic stem cell transplantation; ATG: antithymocyte

globulin; ALG: antilymphocyte globulin; CsA: cyclosporin A; BM: bone marrow; PB: peripheral blood; MSC: mesenchymal stem cell; aGVHD: acute graft-versus-host disease; cGVHD:

chronic graft-versus-host disease; GFFS: graft-versus-host disease-free, failure-free survival; HR: hazard ratio; CI: confidence interval. Values of p<0.05 were considered to be

significant.

conditioning regimens and protocols for GVHD prophylaxis.

No significant differences were observed between the MSC

and non-MSC groups in the present study in terms of EBVassociated

PTLD, pulmonary infections, septicemia, and other

non-infection complications. Thus, our data indicate that MSC

applications are safe.

The rate of TRM in the MSC group was 21.7%, comparable to

the rate observed in the non-MSC group (27.2%, p=0.424).

Infection, GF, and GVHD were the most common causes of

death. We also observed comparable rates of OS between

the two groups (78.3±6.1% vs. 70.1±6.3%, p=0.292), with GF

being the main adverse factor. These findings were consistent

with previous reports [12,31]. Surprisingly, we found that

the estimated 5-year GFFS was significantly improved in the

MSC group compared to the non-MSC group (76.6±6.2% vs.

56.7±6.9%, p=0.045). Possible explanations for this finding

include the MSCs promoting faster engraftment to reduce

the chance of infection or MSCs reducing the risk of cGVHD

to improve the quality of life. GF, grade III-IV aGVHD, and

moderate-severe cGVHD were the factors that predicted worse

126


Turk J Hematol 2022;39:117-129

Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

GFFS. These findings suggest the need to reduce GF and GVHD,

which may improve survival outcomes. Co-transplantation of

MSCs during HSCT is a good attempt in this direction, although

some results were not satisfactory.

Our study has several limitations. First, certain shortcomings are

inherent in retrospective, single-center studies. A prospective,

multicenter, head-to-head clinical trial should be performed

to confirm our results. Second, other factors that may affect

engraftment and GVHD, such as variables of the patient

populations and conditioning regimens, should be controlled in

future studies. Third, many factors may influence the effects

of MSC treatment, such as the source of the MSCs (BM-MSCs

versus UC-MSCs), the frequency of MSC infusions (once,

twice, or more), therapeutic schedules, and the timing of MSC

administrations. All of these factors require more studies to

explore their impacts on efficacy. Despite these limitations,

however, our study offers remarkable comparative evidence

of the value of the prophylactic use of MSCs in haplo-HSCT

treatment for SAA patients.

Conclusion

The work presented here is the first head-to-head, retrospective

study to date to explore the efficacy and safety of the cotransplantation

of UC-MSCs during haplo-HSCT for the

treatment of patients with SAA. The encouraging results

suggest that infusion of MSCs after haplo-HSCT can promote

engraftment, reduce the incidence of cGVHD, and improve

GFFS. The prophylactic use of MSCs combined with haplo-HSCT

was found to be an effective and safe method for the treatment

of patients with SAA.

Acknowledgments: This work was supported by the Natural

Science Foundation of Zhejiang Province (No. LY19H290003), the

Zhejiang Provincial Medical and Health Science and Technology

Project (Nos. 2020KY196, 2018277310), the Foundation of

Zhejiang Province Chinese Medicine Science and Technology

Planes (Nos. 2017ZB030, 2020ZA044), and the Key Project of

the 2017 School Research Fund of Zhejiang Chinese Medical

University (No. 2017ZZ02).

Ethics

Ethics Committee Approval: This study was approved by the

Ethics Committee of the First Affiliated Hospital of Zhejiang

Chinese Medical University.

Informed Consent: All patients signed informed consent forms

in accordance with the Declaration of Helsinki.

Authorship Contributions

Surgical and Medical Practices: H-F.Z., X-F.S., H.L., L-L.H.;

Concept: H-F.Z.; Design: H-F.Z.; Data Collection or Processing:

H-F.Z., X-F.S., H.L., L-L.H.; Analysis or Interpretation: X-F.S., H.L.,

L-L.H.; Writing: X-F.S., H.L., L-L.H.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Zhu XF, He HL, Wang SQ, Tang JY, Han B, Zhang DH, Wu LQ, Wu DP, Li W, Xia

LH, Zhu HL, Liu F, Shi HX, Zhang X, Zhou F, Hu JD, Fang JP, Chen XQ, Ye TZ,

Liang YM, Jin J, Zhang FK. Current treatment patterns of aplastic anemia in

China: a prospective cohort registry study. Acta Haematol 2019;142:162-

170.

2. ElGohary G, El Fakih R, de Latour R, Risitano A, Marsh J, Schrezenmeier H,

Gluckman E, Hochsmann B, Pierri F, Halkes C, Alzahrani H, De la Fuente

J, Cesaro S, Alahmari A, Ahmed SO, Passweg J, Dufour C, Bacigalupo A,

Aljurf M. Haploidentical hematopoietic stem cell transplantation in aplastic

anemia: a systematic review and meta-analysis of clinical outcome on

behalf of the severe aplastic anemia working party of the European group

for blood and marrow transplantation (SAAWP of EBMT). Bone Marrow

Transplant 2020;55:1906-1917.

3. Killick SB, Bown N, Cavenagh J, Dokal I, Foukaneli T, Hill A, Hillmen P,

Ireland R, Kulasekararaj A, Mufti G, Snowden JA, Samarasinghe S, Wood

A, Marsh JC; British Society for Standards in Haematology. Guidelines for

the diagnosis and management of adult aplastic anaemia. Br J Haematol

2016;172:187-207.

4. Bacigalupo A, Giammarco S, Sica S. Bone marrow transplantation versus

immunosuppressive therapy in patients with acquired severe aplastic

anemia. Int J Hematol 2016;104:168-174.

5. Xu LP, Zhang XH, Wang FR, Mo XD, Han TT, Han W, Chen YH, Zhang YY,

Wang JZ, Yan CH, Sun YQ, Zuo SN, Huang XJ. Haploidentical transplantation

for pediatric patients with acquired severe aplastic anemia. Bone Marrow

Transplant 2017;52:381-387.

6. Xu LP, Jin S, Wang SQ, Xia LH, Bai H, Gao SJ, Liu QF, Wang JM, Wang X,

Jiang M, Zhang X, Wu DP, Huang XJ. Upfront haploidentical transplant for

acquired severe aplastic anemia: registry-based comparison with matched

related transplant. J Hematol Oncol 2017;10:25.

7. Ye L, Zhang F, Kojima S. Current insights into the treatments of severe

aplastic anemia in China. Int J Hematol 2020;112:292-299.

8. Dufour C, Veys P, Carraro E, Bhatnagar N, Pillon M, Wynn R, Gibson B, Vora

AJ, Steward CG, Ewins AM, Hough RE, de la Fuente J, Velangi M, Amrolia PJ,

Skinner R, Bacigalupo A, Risitano AM, Socie G, Peffault de Latour R, Passweg

J, Rovo A, Tichelli A, Schrezenmeier H, Hochsmann B, Bader P, van Biezen

A, Aljurf MD, Kulasekararaj A, Marsh JC, Samarasinghe S. Similar outcome

of upfront-unrelated and matched sibling stem cell transplantation

in idiopathic paediatric aplastic anaemia. A study on behalf of the UK

Paediatric BMT Working Party, Paediatric Diseases Working Party and Severe

Aplastic Anaemia Working Party of EBMT. Br J Haematol 2015;171:585-594.

9. Yue C, Ding Y, Gao Y, Li L, Pang Y, Liu Z, Zhang H, Xiao Y, Jiang Z, Xiao

H. Cotransplantation of haploidentical hematopoietic stem cells and

allogeneic bone marrow-derived mesenchymal stromal cells as a first-line

treatment in very severe aplastic anemia patients with refractory infections.

Eur J Haematol 2018;100:624-629.

10. Zeng Y, Wang S, Wang J, Liu L, Su Y, Lu Z, Zhang X, Zhang Y, Zhong JF, Peng

L, Liu Q, Lu Y, Gao L, Zhang X. Optimal donor for severe aplastic anemia

patient requiring allogeneic hematopoietic stem cell transplantation: a

large-sample study from China. Sci Rep 2018;8:2479.

11. Xu L, Liu Z, Wu Y, Yang X, Cao Y, Li X, Yan B, Li S, Da W, Wu X. Clinical

evaluation of haploidentical hematopoietic combined with human

127


Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

Turk J Hematol 2022;39:117-129

umbilical cord-derived mesenchymal stem cells in severe aplastic anemia.

Eur J Med Res 2018;23:12.

12. Liu Z, Wu X, Wang S, Xia L, Xiao H, Li Y, Li H, Zhang Y, Xu D, Nie D, Lai Y, Wu B,

Lin D, Du X, Jiang Z, Gao Y, Gu X, Xiao Y. Co-transplantation of mesenchymal

stem cells makes haploidentical HSCT a potential comparable therapy with

matched sibling donor HSCT for patients with severe aplastic anemia. Ther

Adv Hematol 2020;11:2040620720965411.

13. Liu Z, Zhang Y, Xiao H, Yao Z, Zhang H, Liu Q, Wu B, Nie D, Li Y, Pang Y, Fan Z,

Li L, Jiang Z, Duan F, Li H, Zhang P, Gao Y, Ouyang L, Yue C, Xie M, Shi C, Xiao

Y, Wang S. Cotransplantation of bone marrow-derived mesenchymal stem

cells in haploidentical hematopoietic stem cell transplantation in patients

with severe aplastic anemia: an interim summary for a multicenter phase II

trial results. Bone Marrow Transplant 2017;52:704-710.

14. Wang Z, Yu H, Cao F, Liu Z, Liu Z, Feng W, Liu X, Yu Y, Xiao Y, Li L, Zhou

J. Donor-derived marrow mesenchymal stromal cell co-transplantation

following a haploidentical hematopoietic stem cell transplantation trail to

treat severe aplastic anemia in children. Ann Hematol 2019;98:473-479.

15. Ding L, Han DM, Zheng XL, Yan HM, Xue M, Liu J, Zhu L, Li S, Mao N, Guo

ZK, Ning HM, Wang HX, Zhu H. A study of human leukocyte antigenhaploidentical

hematopoietic stem cells transplantation combined with

allogenic mesenchymal stem cell infusion for treatment of severe aplastic

anemia in pediatric and adolescent patients. Stem Cells Transl Med

2021;10:291-302.

16. Li R, Tu J, Zhao J, Pan H, Fang L, Shi J. Mesenchymal stromal cells as

prophylaxis for graft-versus-host disease in haplo-identical hematopoietic

stem cell transplantation recipients with severe aplastic anemia?-a

systematic review and meta-analysis. Stem Cell Res Ther 2021;12:106.

17. Cui Q, Sha P, Chen H, Shen H, Qin L, Li Z, Wu T, Wang Z. Modified

immunosuppressive therapy with porcine antilymphocyte globulin plus

delayed cyclosporine A in children with severe aplastic anemia. Int J

Hematol 2018;107:64-68.

18. Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P, Hows J,

Thomas ED. 1994 Consensus Conference on Acute GVHD Grading. Bone

Marrow Transplant 1995;15:825-828.

19. Jagasia MH, Greinix HT, Arora M, Williams KM, Wolff D, Cowen EW, Palmer J,

Weisdorf D, Treister NS, Cheng GS, Kerr H, Stratton P, Duarte RF, McDonald

GB, Inamoto Y, Vigorito A, Arai S, Datiles MB, Jacobsohn D, Heller T, Kitko

CL, Mitchell SA, Martin PJ, Shulman H, Wu RS, Cutler CS, Vogelsang GB,

Lee SJ, Pavletic SZ, Flowers ME. National Institutes of Health Consensus

Development Project on Criteria for Clinical Trials in Chronic Graft-versus-

Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol

Blood Marrow Transplant 2015;21:389-401.e1.

20. Xu ZL, Zhou M, Jia JS, Mo WJ, Zhang XH, Zhang YP, Wang Y, Li YM, Huang

XJ, Wang SQ, Xu LP. Immunosuppressive therapy versus haploidentical

transplantation in adults with acquired severe aplastic anemia. Bone

Marrow Transplant 2019;54:1319-1326.

21. Zhang Y, Guo Z, Liu XD, He XP, Yang K, Chen P, Chen HR. Comparison

of haploidentical hematopoietic stem cell transplantation and

immunosuppressive therapy for the treatment of acquired severe aplastic

anemia in pediatric patients. Am J Ther 2017;24:e196-e201.

22. Huang XJ. HLA-mismatched/haploidentical hematopoietic stem cell

transplantation: a field in which Chinese doctors are making great

contributions. Chin Med J (Engl) 2010;123:1235-1240.

23. Ciceri F, Lupo-Stanghellini MT, Korthof ET. Haploidentical transplantation

in patients with acquired aplastic anemia. Bone Marrow Transplant

2013;48:183-185.

24. Liu LM, Zhang YM, Zhou HF, Wang QY, Qiu HY, Tang XW, Han Y, Fu CC, Jin ZM,

Sun AN, Miao M, Wu DP. Outcome of combination of HLA-haploidentical

hematopoietic SCT with an unrelated cord blood unit for 127 patients with

acquired severe aplastic anemia. Zhonghua Xue Ye Xue Za Zhi 2018;39:624-

628.

25. Zhao K, Liu Q. The clinical application of mesenchymal stromal cells in

hematopoietic stem cell transplantation. J Hematol Oncol 2016;9:46.

26. Wang H, Wang Z, Xue M, Liu J, Yan H, Guo Z. Co-transfusion of haploidentical

hematopoietic and mesenchymal stromal cells to treat a patient

with severe aplastic. Cytotherapy 2010;12:563-565.

27. Wu Y, Cao Y, Li X, Xu L, Wang Z, Liu P, Yan P, Liu Z, Wang J, Jiang S, Wu X,

Gao C, Da W, Han Z. Cotransplantation of haploidentical hematopoietic

and umbilical cord mesenchymal stem cells for severe aplastic anemia:

successful engraftment and mild GVHD. Stem Cell Res 2014;12:132-138.

28. Wagner JL, Deeg HJ, Seidel K, Anasetti C, Doney K, Sanders J, Sullivan

KM, Storb R. Bone marrow transplantation for severe aplastic anemia

from genotypically HLA-nonidentical relatives. An update of the Seattle

experience. Transplantation 1996;61:54-61.

29. Xu LP, Liu KY, Liu DH, Han W, Chen H, Chen YH, Zhang XH, Wang Y, Wang FR,

Wang JZ, Huang XJ. A novel protocol for haploidentical hematopoietic SCT

without in vitro T-cell depletion in the treatment of severe acquired aplastic

anemia. Bone Marrow Transplant 2012;47:1507-1512.

30. Xu LP, Wang SQ, Wu DP, Wang JM, Gao SJ, Jiang M, Wang CB, Zhang X, Liu

QF, Xia LH, Wang X, Huang XJ. Haplo-identical transplantation for acquired

severe aplastic anaemia in a multicentre prospective study. Br J Haematol

2016;175:265-274.

31. Li Y, Duan F, Xiao H, Wu X, Wang S, Xu D, Liu Q, Fan Z, Nie D, Lai Y, Wu B, Lin D,

Du X, Weng J, Jiang Z, Pang Y, Ouyang L, Liu Z, Zhang L, Han N, Chen L, Xiao

Y. Therapeutic outcomes of haploidentical allogeneic hematopoietic stem

cell transplantation in patients with severe aplastic anemia: a multicenter

study. Transplantation 2018;102:1724-1731.

32. Liu L, Zhang Y, Jiao W, Zhou H, Wang Q, Qiu H, Tang X, Han Y, Fu C, Jin Z, Chen

S, Sun A, Miao M, Wu D. Combination of haploidentical haematopoietic

stem cell transplantation with an unrelated cord-blood unit in patients

with severe aplastic anemia: a report of 146 cases. Bone Marrow Transplant

2020;55:2017-2025.

33. Kuzmina LA, Petinati NA, Vasilieva VA, Dovydenko MV, Drokov MY, Davydova

YO, Kapranov NM, Sats NV, Chabaeva YA, Kulikov SM, Gaponova TV, Drize

NI, Parovichnikova EN, Savchenko VG. Multipotent mesenchymal stromal

cells application for acute graft versus host disease treatment. Ter Arkh

2020;92:23-30.

34. Kurtzberg J, Abdel-Azim H, Carpenter P, Chaudhury S, Horn B, Mahadeo K,

Nemecek E, Neudorf S, Prasad V, Prockop S, Quigg T, Satwani P, Cheng A,

Burke E, Hayes J, Skerrett D; MSB-GVHD001/002 Study Group. A phase 3,

single-arm, prospective study of remestemcel-L, ex vivo culture-expanded

adult human mesenchymal stromal cells for the treatment of pediatric

patients who failed to respond to steroid treatment for acute graft-versushost

disease. Biol Blood Marrow Transplant 2020;26:845-854.

35. Zhao L, Chen S, Yang P, Cao H, Li L. The role of mesenchymal stem cells

in hematopoietic stem cell transplantation: prevention and treatment of

graft-versus-host disease. Stem Cell Res Ther 2019;10:182.

36. Kuzmina LA, Petinati NA, Parovichnikova EN, Lubimova LS, Gribanova EO,

Gaponova TV, Shipounova IN, Zhironkina OA, Bigildeev AE, Svinareva DA,

Drize NJ, Savchenko VG. Multipotent mesenchymal stromal cells for the

prophylaxis of acute graft-versus-host disease-a phase II study. Stem Cells

Int 2012;2012:968213.

37. Gao L, Zhang Y, Hu B, Liu J, Kong P, Lou S, Su Y, Yang T, Li H, Liu Y, Zhang C,

Gao L, Zhu L, Wen Q, Wang P, Chen X, Zhong J, Zhang X. Phase II multicenter,

randomized, double-blind controlled study of efficacy and safety of

umbilical cord-derived mesenchymal stromal cells in the prophylaxis of

chronic graft-versus-host disease after HLA-haploidentical stem-cell

transplantation. J Clin Oncol 2016;34:2843-2850.

38. Kogler G, Radke TF, Lefort A, Sensken S, Fischer J, Sorg RV, Wernet P.

Cytokine production and hematopoiesis supporting activity of cord bloodderived

unrestricted somatic stem cells. Exp Hematol 2005;33:573-583.

128


Turk J Hematol 2022;39:117-129

Sheng X-F. et al: Combination of Haplo-HSCT with UC-MSCs

39. Forslow U, Blennow O, LeBlanc K, Ringden O, Gustafsson B, Mattsson J,

Remberger M. Treatment with mesenchymal stromal cells is a risk factor

for pneumonia-related death after allogeneic hematopoietic stem cell

transplantation. Eur J Haematol 2012;89:220-227.

40. Zu Y, Zhou J, Fu Y, Fang B, Liu X, Zhang Y, Yu F, Zuo W, Zhou H, Gui R, Li Z,

Liu Y, Zhao H, Zhang C, Song Y. Feasibility of reduced-dose posttransplant

cyclophosphamide and cotransplantation of peripheral blood stem cells

and umbilical cord-derived mesenchymal stem cells for SAA. Sci Rep

2021;11:253.

129


RESEARCH ARTICLE

DOI: 10.4274/tjh.galenos.2022.2021.0670

Turk J Hematol 2022;39:130-135

Castleman Disease: A Multicenter Case Series from Turkey

Castleman Hastalığı: Türkiye’den Çok Merkezli Bir Olgu Serisi

Eren Gündüz 1 , Hakkı Onur Kırkızlar 2 , Elif Gülsüm Ümit 2 , Sedanur Karaman Gülsaran 2 , Vildan Özkocaman 3 ,

Fahir Özkalemkaş 3 , Ömer Candar 3 , Tugrul Elverdi 4 , Selin Küçükyurt 4 , Semra Paydaş 5 , Özcan Çeneli 6 , Sema Karakuş 7 ,

Senem Maral 8 , Ömer Ekinci 9 , Yıldız İpek 10 , Cem Kis 11 , Zeynep Tuğba Güven 12 , Aydan Akdeniz 13 , Tiraje Celkan 14 ,

Ayşe Hilal Eroğlu Küçükdiler 15 , Gülsüm Akgün Çağlıyan 16 , Ceyda Özçelik Şengöz 17 , Ayşe Karataş 18 , Tuba Bulduk 19 ,

Alper Özcan 12 , Fatma Burcu Belen Apak 7 , Aylin Canbolat 20 , İbrahim Kartal 21 , Hale Ören 22 , Ersin Töret 1 ,

Gül Nihal Özdemir 14 , Şule Mine Bakanay Öztürk 23

1Eskişehir Osmangazi University Faculty of Medicine, Department of Hematology, Eskişehir, Turkey

2Trakya University Faculty of Medicine, Department of Hematology, Edirne, Turkey

3Uludağ University Faculty of Medicine, Department of Hematology, Bursa, Turkey

4İstanbul University-Cerrahpaşa Cerrahpaşa Faculty of Medicine, Department of Hematology, İstanbul, Turkey

5Çukurova University Faculty of Medicine, Department of Hematology, Adana, Turkey

6Necmettin Erbakan University Meram Faculty of Medicine, Department of Hematology, Konya, Turkey

7Ankara Başkent University Faculty of Medicine, Department of Hematology, Ankara, Turkey

8University of Health Sciences Turkey Ankara Dışkapı Yıldırım Beyazıt Training and Research Hospital, Clinic of Hematology, Ankara, Turkey

9Fırat University Faculty of Medicine, Department of Hematology, Elazığ, Turkey

10Kartal Dr. Lütfi Kırdar City Hospital, Clinic of Hematology, İstanbul, Turkey

11Adana Başkent University Faculty of Medicine, Department of Hematology, Adana, Turkey

12Erciyes University Faculty of Medicine, Department of Hematology, Kayseri, Turkey

13Mersin University Faculty of Medicine, Department of Hematology, Mersin, Turkey

14İstinye University Faculty of Medicine, Department of Pediatric Hematology, İstanbul, Turkey

15Adnan Menderes University Faculty of Medicine, Department of Hematology, Aydın, Turkey

16Pamukkale University Faculty of Medicine, Department of Hematology, Denizli, Turkey

17Karadeniz Technical University Faculty of Medicine, Department of Hematology, Trabzon, Turkey

18Hacettepe University Faculty of Medicine, Department of Hematology, Ankara, Turkey

19University of Health Sciences Turkey Gülhane Training and Research Hospital, Clinic of Hematology, Ankara, Turkey

20İstanbul Medeniyet University Faculty of Medicine, Department of Pediatric Hematology, İstanbul, Turkey

21Ondokuz Mayıs University Faculty of Medicine, Department of Pediatric Hematology, Samsun, Turkey

22Dokuz Eylül University Faculty of Medicine, Department of Pediatric Hematology, İzmir, Turkey

23Ankara Yıldırım Beyazıt University Faculty of Medicine, Department of Hematology, Ankara, Turkey

Abstract

Objective: Castleman disease (CD) is a rare disease also known as

angiofollicular lymph node hyperplasia. The two main histological

subtypes are the hyaline vascular and plasma cell variants. It is further

classified as unicentric CD (UCD) or multicentric CD (MCD) according

to the anatomical distribution of the disease and the number of lymph

nodes involved. The aim of this multicenter study was to evaluate all

cases of CD identified to date in Turkey to set up a national registry to

improve the early recognition, treatment, and follow-up of CD.

Öz

Amaç: Anjiyofolliküler lenf nodu hiperplazisi olarak da bilinen

Castleman hastalığı (CH), nadir bir hastalık olup başlıca hiyalin vasküler

ve plazma hücreli olmak üzere 2 histolojik alt tipi vardır. Hastalığın

anatomik yayılımı ve tutulan lenf nodu bölgelerinin sayısına göre

unisentrik (UCH) ya da multisentrik (MCH) olarak sınıflandırılır. Bu

çok merkezli çalışmanın amacı bugüne kadar Türkiye’de tanımlanan

tüm CH olgularını tanımlamak, ulusal bir veri tabanı oluşturarak CH’de

erken tanı, tedavi ve takip sürecine katkı sağlamaktır.

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Eren Gündüz, M.D., Eskişehir Osmangazi University Faculty of Medicine, Received/Geliş tarihi: December 6, 2021

Department of Hematology, Eskişehir, Turkey

Accepted/Kabul tarihi: February 7, 2022

E-mail : erengunduz26@gmail.com ORCID: orcid.org/0000-0002-7660-4092

130


Turk J Hematol 2022;39:130-135

Gündüz E. et al: Case Series of Castleman Disease

Abstract

Materials and Methods: Both adult (n=130) and pediatric (n=10)

patients with lymph node or involved field biopsy results reported as

CD were included in the study. Patients’ demographic information,

clinical and laboratory characteristics, imaging study results, treatment

strategies, and clinical outcomes were evaluated retrospectively.

Results: A total of 140 patients (69 male and 71 female) with a

diagnosis of UCD (n=73) or MCD (n=67) were included. The mean

age was 39 years in the UCD group and 47 years in the MCD group.

Female patients were more common in the UCD group. The most

common histological subtype was hyaline vascular for both UCD

and MCD patients. Asymptomatic patients were more common in

the UCD group. Anemia, elevations of acute phase reactants, and

hypoalbuminemia were more common in the MCD group. The most

commonly used treatment strategies for UCD were surgical excision,

rituximab, and radiotherapy, respectively. All UCD patients were alive

at a median of 19.5 months of follow-up. The most commonly used

treatment strategies for MCD were methyl prednisolone, R-CHOP,

R-CVP, and rituximab. Thirteen MCD patients had died at a median of

34 months of follow-up.

Conclusion: This study is important in presenting the patient

characteristics and treatment strategies for CD from Turkey, with the

potential of increasing awareness about CD. Treatment data may help

in making decisions, particularly in countries that do not have access

to siltuximab. However, larger prospective studies are needed to make

definitive conclusions.

Keywords: Castleman disease, Unicentric, Multicentric, Treatment

Öz

Gereç ve Yöntemler: Çalışmaya lenf nodu ya da tutulan alandan

yapılmış biyopsi sonucu CH olarak rapor edilen hem erişkin

(n=130) hem de pediyatrik (n=10) hastalar dahil edildi. Hastaların

demografik bilgileri, klinik ve laboratuvar özellikleri, görüntüleme

bulguları, aldıkları tedaviler ve tedavi sonuçları geriye dönük olarak

değerlendirildi.

Bulgular: Dahil edilen 140 hastanın 69’u kadın, 71’i erkekti. Yetmiş

üç hasta UCH, 67 hasta MCH olarak sınıflandırılmıştı. Yaş ortalaması

UCH’de 39, MCH’de 47 yıl idi. Kadın hastalar UCH’de daha fazlaydı.

Hem UCH hem de MCH için en sık histolojik alt tip hiyalin vaskülerdi.

Asemptomatik hastalar UCH’de daha fazlaydı. Anemi, akut faz

reaktanı yüksekliği ve hipoalbuminemi MCH’de daha sıktı. UCH’de

en sık uygulanan tedaviler sırasıyla cerrahi eksizyon, rituksimab

ve radyoterapiydi. Median 19,5 aylık takipte tüm UCH’li hastalar

hayattaydı. MCH’de 1. basamak tedaviler metil prednizolon, R-CHOP,

R-CVP ve rituksimab idi. Median 34 aylık takipte 13 MCH’li hasta

kaybedilmişti.

Sonuç: Çalışmamız Türkiye’deki CH hastalarının özellikleri ve tedavi

yaklaşımlarını yansıtması açısından önemli olup hastalıkla ilgili

farkındalığın arttırılması potansiyeline sahiptir. Tedavi verileri özellikle

ülkemizde olduğu gibi siltuksimaba ulaşımı zor olan ülkelerde tedavi

seçimi konusunda fikir verebilir. Kesin sonuçlar çıkarmak için büyük

ölçekli prospektif çalışmalara ihtiyaç vardır.

Anahtar Sözcükler: Castleman hastalığı, Unisentrik, Multisentrik,

Tedavi

Introduction

Castleman disease (CD), also known as angiofollicular lymph

node hyperplasia or giant lymph node hyperplasia, was first

described in 1954 [1,2,3,4]. The two main histological subtypes

are the hyaline vascular and plasma cell variants, and a mixed

variant is seen occasionally [5]. It is further classified as

unicentric CD (UCD) or multicentric CD (MCD) according to the

anatomical distribution of the disease and the number of lymph

nodes involved [6,7]. The estimated incidence is approximately

25 cases per million person-years, which represents fewer than

5200 cases per year in the United States [8,9].

Classically, MCD presents with lymphadenopathy affecting

multiple lymph node stations and is associated with systemic

symptoms such as fever, weight loss, and fatigue, driven by

interleukin-6 and other cytokines. MCD has been subclassified

into human herpesvirus-8 (HHV-8)-associated MCD;

polyneuropathy, organomegaly, endocrinopathy, monoclonal

plasma cell disorder, skin changes (POEMS)-associated MCD; and

idiopathic MCD (iMCD). UCD, on the other hand, involves a single

enlarged lymph node or multiple enlarged lymph nodes within

a single lymph node region. The diagnosis of UCD is frequently

incidental and the lymphadenopathy is often asymptomatic.

However, some patients present with symptoms resulting from

compression of vital structures (e.g., the airways, neurovascular

bundles, or ureters), whereas others will experience iMCDlike

inflammatory syndromes. UCD is virtually always HHV-8-

negative, but rare positive cases have been reported [6].

The aim of this multicenter study is to evaluate all cases of

CD identified to date in Turkey to set up a national registry to

improve the early recognition, treatment, and follow-up of this

disease.

Materials and Methods

We included all patients with a diagnosis of CD based on the

histopathological analysis of a lymph node or other affected area.

Information about the patients was collected retrospectively

and included patients’ demographic information, treatment

strategy, clinical outcome, and the results of laboratory and

imaging studies. Data were collected between April 2018 and

August 2020 and the dates of diagnoses were between 2002

and 2020.

The study was approved by the local ethics committee of

Eskişehir Osmangazi University.

Statistical Analysis

We stratified our patient population based on centricity and

compared baseline clinical characteristics. Categorical variables

131


Gündüz E. et al: Case Series of Castleman Disease

Turk J Hematol 2022;39:130-135

are reported as counts and percentages; parametric continuous

variables are reported as means and standard deviations.

Nonparametric continuous variables are reported as medians

and interquartile ranges. To determine differences between

cohorts for categorical variables, we used chi-square and

Fisher exact tests. To determine differences for nonparametric

continuous variables, we used the Mann-Whitney U test. Finally,

to determine differences for parametric continuous variables,

we used the Student t-test. We also constructed Kaplan-Meier

survival curves for patients with UCD and MCD. All p-values are

two-tailed with a significance level of 0.05 reflecting statistical

significance. We performed the statistical analysis using IBM

SPSS Statistics 21.0 for Windows (IBM Corp., Armonk, NY, USA).

Results

A total of 140 patients (69 male and 71 female) from 21 centers

with a diagnosis of UCD (n=73) or MCD (n=67) were included

in the study. Patients were younger and female patients were

more common in the UCD group than the MCD group. The most

common histological type by lymph node biopsy was hyaline

vascular. Symptoms at diagnosis were similar between the

two groups except for fatigue, anorexia, fever, diarrhea, and

affected inguinal lymph nodes. Asymptomatic patients were

more common in the UCD group.

Physical examinations revealed significant differences between

the presence of hepatomegaly, splenomegaly, and affected

submandibular, axillary, and inguinal lymph nodes in the UCD

and MCD groups. Affected lymph nodes in the UCD group

were most frequently found in the submandibular region (n=5,

17.2%) (Table 1).

Serum C-reactive protein (CRP) level was increased in 15 (36.6%)

patients and elevated immunoglobulin (Ig) G was observed in 6

(18.8%) patients.

Imaging studies performed for patients with UCD included neck

computed tomography (CT) for 39 (56.5%), thorax CT for 43

(50%), abdominopelvic CT for 38 (46.9%), and positron emission

tomography (PET)/CT for 31 (44.3%) patients. Lymphadenopathy

was observed in the cervical area for 19 (44.2%), intrathoracic

area for 8 (20.5%), and abdominopelvic area for 14 (28.6%)

patients (Table 2).

Table 1. Clinical characteristics of the patients.

132

Unicentric (n=73) Multicentric (n=67) p

Age at diagnosis (years) 38.95±16.06 48.61±19.45 0.002

Sex ratio (female/male) 45/28 26/41 0.009

Histological subtype

Hyaline vascular

Plasma cell

Mixed

Unknown

Most frequent symptoms at diagnosis

Fatigue

Anorexia

Fever

Weight loss

Sweating

Cough

Diarrhea

Cervical lymph node involvement

Axillary lymph node involvement

Inguinal lymph node involvement

Abdominal pain

Physical examination

Hepatomegaly

Splenomegaly

Submandibular lymph node involvement

Submental lymph node involvement

Supraclavicular lymph node involvement

Axillary lymph node involvement

Inguinal lymph node involvement

53 (72.6%)

11 (15.1%)

5 (6.8%)

4 (5.5%)

15 (35.7%)

4 (22.2%)

4 (21.1%)

11 (40.7%)

6 (40%)

6 (37.5%)

0 (0%)

22 (53.7%)

6 (31.6%)

2 (16.7%)

19 (66.7%)

7 (25.9%)

3 (9.7%)

5 (17.2%)

1 (14.3%)

4 (26.7%)

7 (15.9%)

3 (7.9%)

26 (37.9%)

22 (33.3%)

5 (7.6%)

14 (21.2%)

27 (64.3%)

14 (77.8%)

15 (78.9%)

16 (59.3%)

9 (60%)

10 (62.5%)

4 (100%)

19 (46.3%)

13 (68.4%)

10 (83.3%)

5 (33.3%)

20 (74.1%)

28 (90.3%)

24 (82.8%)

6 (85.7%)

11 (73.3%)

37 (84.1%)

35 (92.1%)

<0.001

<0.001

<0.001

<0.001

0.015

0.012

0.007

0.266

0.451

0.311

0.048

1.000

0.085

0.021

0.359

0.004

0.012

<0.001

0.053

0.064

<0.001

<0.001


Turk J Hematol 2022;39:130-135

Gündüz E. et al: Case Series of Castleman Disease

The most common first-line treatment in cases of UCD was

surgical excision, followed by rituximab and radiotherapy.

Thirty-nine patients were followed without treatment (Table

3). Twenty-eight (87.5%) patients were in complete remission

and 3 (9.4%) patients were in partial remission after first-line

treatment. Response to first-line treatment was not evaluated

for 2 (6.3%) patients. Only 3 patients needed a second-line

treatment and their treatment responses were complete

remission. Second-line therapies were radiotherapy (n=1),

cyclosporine (n=1), and chemoimmunotherapy (R-ESHAP) (n=1).

At the last evaluation after a median follow-up of 19.5 (range:

7-52.5) months, all patients with UCD were alive (Figure 1).

The most common histological type of CD by lymph node biopsy

was also the hyaline vascular type in cases of MCD. Coronary

artery disease, chronic renal failure, and solid malignancy were

more common in the MCD group, possibly due to the older

mean age of this group. Kaposi sarcoma was reported for two

MCD patients.

The most common symptoms and physical examination findings

were reported as fever (n=15, 78.8%), affected inguinal

lymph nodes (n=10, 83.3%), hepatomegaly (n=20, 74.1%),

splenomegaly (n=28, 90.3%), arthralgia (n=2, 40%), abdominal

pain (n=5, 33.3%), fatigue (n=27, 33.3%), and diarrhea (n=4,

100%). Serum CRP levels were increased in 26 (63.4%) cases.

Elevated IgG levels were detected in 26 (81.3%) patients.

Anemia, thrombocytopenia, elevated erythrocyte sedimentation

rate, and hypoalbuminemia were more common in the MCD

group than the UCD group.

Imaging studies performed for MCD patients included neck

CT for 30 (43.5%), thorax CT for 43 (50%), abdominopelvic CT

for 43 (53.1%), and PET/CT for 39 (55.7%). Lymphadenopathy

was identified in the cervical area for 24 (55.8%) patients, the

intrathoracic area for 31 (79.5%), and the abdominopelvic area

for 35 (71.4%). Most of the affected lymph nodes identified

by thorax and abdominopelvic CT were smaller than 5 cm in

diameter.

SUV max

values in PET/CT were similar between the two groups

and most commonly below 6. However, PET/CT positivity was

more common in the MCD group.

Follow up (months)

Figure 1. Survival curves of patients.

Type of involvement

1: Unicentric Castleman disease, 2: Multicentric Castleman disease.

Table 2. Laboratory characteristics of the patients.

Laboratory findings

Anemia

Lymphopenia

Thrombocytopenia

Elevated ESR

Elevated CRP

Elevated β2 microglobulin

Hypoalbuminemia

Elevated IgG

Elevated IgA

HHV-8 positivity

HIV positivity

Imaging studies

Lymph node in neck CT

Lymph node in thorax CT

Lymph node in abdominopelvic CT

Activity in PET/CT

Unicentric (n=73) Multicentric (n=67) p

19 (34.5%)

8 (50%)

2 (13.3%)

9 (19.1%)

15 (36.6%)

11 (25.6%)

3 (9.1%)

6 (18.8%)

1 (5%)

0 (0%)

0 (0%)

19 (44.2%)

8 (20.5%)

14 (28.6%)

22 (36.1%)

36 (65.5%)

8 (50%)

13 (80.9%)

38 (80.9%)

26 (63.4%)

32 (74.4%)

30 (90.9%)

26 (81.3%)

19 (95%)

13 (100%)

0 (0%)

24 (55.8%)

31 (79.5%)

35 (71.4%)

39 (63.9%)

0.002

0.884

0.003

<0.001

0.048

<0.001

<0.001

<0.001

<0.001

<0.001

1.000

0.012

<0.001

<0.001

<0.001

ESR: Erythrocyte sedimentation rate; CRP: C-reactive protein; IgG: immunoglobulin G; IgA: immunoglobulin A; HHV-8: human herpesvirus-8; HIV: human immunodeficiency virus; CT:

computed tomography; PET: positron emission tomography.

133


Gündüz E. et al: Case Series of Castleman Disease

Turk J Hematol 2022;39:130-135

Patients in the MCD group received methyl prednisolone (n=4,

10.5%) R-CHOP (n=5, 13.2%), R-CVP (n=3, 7.9%), CVP (n=4,

10.5%), CHOP (n=3, 7.9%), and rituximab (n=16, 42.1%) as

first-line treatments (Table 2). Thirteen (37.1%) patients were

in complete remission and 13 (37.1%) patients were in partial

remission after first-line treatment. Five (14.3%) patients had

progressive and 4 (11.4%) patients had stable disease. Response

to first-line treatment was not evaluated for 3 patients in this

group.

More patients in the MCD group needed second-line treatment

compared to the UCD group (18.8% vs. 81.3%, p=0.021).

Second-line treatments included combined chemotherapy (CVP,

CHOP; n=5), chemoimmunotherapy (R-CVP, R-etoposide; n=3),

rituximab (n=1), lenalidomide (n=1), tocilizumab (n=1), and

surgical excision (n=1). Treatment responses in these cases were

progressive disease (n=1), complete remission (n=3), partial

remission (n=6), and stable disease (n=4). Two MCD patients

needed third-line (chemoimmunotherapy, tocilizumab) and 2

patients needed fourth-line (lenalidomide, methyl prednisolone)

treatment. One patient underwent autologous hematopoietic

stem cell transplantation as a fifth-line treatment and had

stable disease at the time of evaluation.

At the last evaluation after a median follow-up of 34 (range:

10-59) months, 13 (34.2%) patients with MCD had died (Figure

1).

Discussion

In this study, our aim was to review the largest cohort and set up

a national registry for a rare disease in Turkey. Another aim of

the study was to increase the awareness about CD and prevent

diagnostic delays.

UCD is reported in approximately 75% of cases of CD [10,11].

In our study, UCD patients accounted for 52% of all patients.

The plasmablastic subtype is different from the three main

histological types and is observed in HHV-8-positive patients

[12]. The plasmablastic subtype was not reported among our

patients, but the subtypes of several patients were not known.

Although Pribyl et al. [5] reported a marginal female

predominance among patients with UCD, CD generally affects

both sexes equally [1,13]. We found a significant female

predominance among our patients with UCD. Patients with

UCD tend to present in the second and fourth decades of life,

being significantly younger than those with MCD, the latter

of which has peak incidence in the sixth and seventh decades

of life [7,13]. Our patients with MCD were older than the UCD

patients but generally younger than MCD patients reported in

the literature.

Systemic symptoms and laboratory abnormalities are more

commonly reported in cases of MCD in the literature [2]. Our

MCD patients also had more systemic symptoms and laboratory

abnormalities than those with UCD.

Viral infections are postulated to play a role in the pathogenesis

of CD; in particular, an association of HHV-8 with MCD is

reported [14,15,16,17]. In a meta-analysis conducted by Talat

et al. [2], HHV-8 was reported to be positive in 46 of 49 (93.9%)

patients with MCD. HHV-8 was evaluated for 39 of our MCD

patients and was found to be positive in 13 (33.3%) cases.

Coinfections of HHV-8 and HIV are also commonly observed

[14], but none of our patients were HIV-positive.

Upon histopathological examination, UCD predominantly

consists of the hyaline vascular variant (90%), while in MCD the

plasmacytoid variant is most commonly observed [16,18,19]. The

hyaline vascular variant was the most common histopathological

subtype in both our UCD and MCD groups.

PET/CT is the suggested imaging method if it can be performed

[20]. Maximum SUV max

values are reported as ranging from 3

to 8 in cases of CD and lymphoma should be suspected in the

differential diagnosis if the value is above 8 [21,22]. In our study,

PET/CT SUV max

values were between 2.5 and 5.

MCD is a rare disease and no prospective randomized controlled

trials have been performed. Therefore, treatment strategies

are heterogeneous, particularly in cases of iMCD [23]. This

heterogeneity was also observed in our study and our data were

Table 3. First-line treatments of patients.

First-line treatment

R-CHOP

Rituximab

R-CVP

Surgical excision

Methyl prednisolone

CVP

CHOP

Radiotherapy

Unicentric (n=34)

1 (2.9%)

2 (5.9%)

0 (0%)

28 (82.4%)

1 (2.9%)

0 (0%)

0 (0%)

2 (5.9%)

Multicentric (n=38)

5 (13.2%)

16 (42.1%)

3 (7.9%)

2 (5.3%)

4 (10.5%)

4 (10.5%)

3 (7.9%)

1 (2.6%)

R: Rituximab; CHOP: cyclophosphamide, doxorubicin, vincristine, methyl prednisolone; CVP: cyclophosphamide, vincristine, methyl prednisolone.

134


Turk J Hematol 2022;39:130-135

Gündüz E. et al: Case Series of Castleman Disease

not sufficient for definitive conclusions. Although siltuximab is

commonly suggested as a first-line treatment, no experiences

with siltuximab were reported in our study because we are

unable to access this drug due to reimbursement obstacles in

Turkey. Central pathological revision was not performed and

this may be another limitation of our study.

Conclusion

We have reported the results of a multicenter retrospective study

of patients with CD, which is a rare disease. The data reported

here are important as they represent the patient characteristics

and treatment strategies from Turkey and have the potential of

increasing awareness about CD. Such treatment data may also

help in making decisions, particularly in countries that do not

have access to siltuximab. However, larger prospective studies

are needed to draw definitive conclusions regarding optimal

treatment options.

Acknowledgment: We would like to thank Associate Professor

Cengiz Bal from the Eskişehir Osmangazi University Faculty of

Medicine’s Department of Biostatistics for statistical analysis.

Ethics

Ethics Committee Approval: The study was approved by the

local ethics committee of Eskişehir Osmangazi University.

Informed Consent: Retrospective study.

Authorship Contributions

Surgical and Medical Practices: E.Ö., H.O.K., E.G.Ü., S.K.G., V.Ö.,

F.Ö., Ö.C., T.E., S.Kü., S.P., Ö.Ç., S.Ka., S.M., Ö.E., Y.İ., C.K., Z.T.G.,

A.A., T.C., A.E.H.K., G.A.Ç., C.Ö.Ş., A.K., T.B., A.Ö., F.B.B.A., A.C.,

İ.K., H.Ö., E.T., G.N.Ö., Ş.M.B.Ö.; Concept: E.G., S.Kü., G.N.Ö.,

Ş.M.B.Ö.; Design: E.G., S.Kü., G.N.Ö., Ş.M.B.Ö.; Data Collection

or Processing: E.G.; Analysis or Interpretation: E.G.; Literature

Search: E.G.; Writing: E.G.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The database project of this study was

supported by the Turkish Society of Hematology.

References

1. Kim MH, Hwang S, Choi YB, Oh ST, Kim SC, Choi GM, Ahn CS, Kim KH, Moon

DB, Ha TY, Song GW, Jung DH, Yu ES, Lee SG. Castleman disease of the

abdomen-single-center experience of 13 surgically treated patients over 11

years. Hepatogastroenterology 2010;57:1060-1063.

2. Talat N, Belgaumkar AP, Schulte KM. Surgery in Castleman’s disease: a

systematic review of 404 published cases. Ann Surg 2012;255:677-684.

3. Anagnostou D, Harrison CV. Angiofollicular lymph node hyperplasia

(Castleman). J Clin Pathol 1972;25:306-311.

4. Chuwa WLE, Chuah KL, Ong HS. Unusual presentation of abdominal

Castleman’s disease. Asian J Surg 2006;29:153-156.

5. Pribyl K, Vakayil V, Farooqi N, Arora N, Kreitz B, Ikramuddin S, Linden MA,

Harmon J. Castleman disease: a single center case series. Int J Surg Case Rep

2021;80:105650.

6. van Rhee F, Oksenhendler E, Srkalovic G, Voorhees P, Lim M, Dispenzieri A,

Ide M, Parente S, Schey S, Streetly M, Wong R, Wu D, Maillard I, Brandstadter

J, Munshi N, Bowne W, Elenitoba-Johnson KS, Fössa A, Lechowicz MJ,

Chandrakasan S, Pierson SK, Greenway A, Nasta S, Yoshizaki K, Kurzrock

R, Uldrick TS, Casper C, Chadburn A, Fajgenbaum DC. International

evidence-based consensus diagnostic and treatment guidelines for

unicentric Castleman disease. Blood Adv 2020;4:6039-6050.

7. Keller AR, Hochholzer L, Castleman B. Hyaline-vascular and plasma-cell

types of giant lymph node hyperplasia of the mediastinum and other

locations. Cancer 1972;29:670-683.

8. Simpson D. Epidemiology of Castleman disease. Hematol Oncol Clin North

Am 2018;32:1-10.

9. Chan KL, Lade S, Prince HM, Harrison SJ. Update and new approaches in the

treatment of Castleman disease. J Blood Med 2016;7:145-158.

10. Wu D, Lim MS, Jaffe ES. Pathology of Castleman disease. Hematol Oncol

Clin North Am 2018;32:37-52.

11. Wong RSM. Unicentric Castleman disease. Hematol Oncol Clin North Am

2018;32:65-73.

12. Wojtyś M, Piekarska A, Kunc M, Ptaszyński K, Biernat W, Zaucha JM,

Waloszczyk P, Lisowski P, Kubisa B, Grodzki T. Clinicopathological

comparison and therapeutic approach to Castleman disease-a case-based

review. J Thorac Dis 2019;11:4859-4874.

13. Puram SV, Hasserjian RP, Faquin WC, Lin HW, Rocco WJ. Castleman disease

presenting in the neck: report of a case and review of the literature. Am J

Otolaringol 2013;34:239-244.

14. Fajgenbaum DC, Shilling D. Castleman disease pathogenesis. Hematol Oncol

Clin North Am 2018; 32:11-21.

15. Soumerai JD, Sohani AR, Abramson JS. Diagnosis and management of

Castleman disease. Cancer Control 2014;21:266-278.

16. Ren N, Ding L, Jia E, Xue J. Recurrence in unicentric Castleman’s disease

postoperatively: a case report and literature review. BMC Surg 2018;18:1.

17. Chronowski GM, Ha CS, Wilder RB, Cabanillas F, Manning J, Cox JD.

Treatment of unicentric and multicentric Castleman disease and the role of

radiotherapy. Cancer 2001; 92:670-676.

18. Wang HW, Pittaluga S, Jaffe ES. Multicentric Castleman disease: where are

we now? Semin Diagn Pathol 2016;33:294-306.

19. Bracale U, Pacelli F, Milone M, Bracale UM, Sodo M, Merola G, Troiani T,

Di Salvo E. Laparoscopic treatment of abdominal unicentric Castleman’s

disease: a case report and literature review. BMC Surg 2017;17:38.

20. Slazat R, Munchi NC. Diagnosis of Castleman disease. Hematol Oncol Clin

N Am 2018;32:53-64.

21. Koa B, Borja AJ, Aly M, Padmanabhan S, Tran J, Zhang V, Rojulpote C, Pierson

SK, Tamakloe MA, Khor JS, Werner TJ, Fajgenbaum DC, Alavi A, Revheim ME.

Emerging role of 18F-FDG PET/CT in Castleman disease: a review. Insights

Imaging 2021;12:35.

22. Oksenhendler E, Boutboul D, Fajgenbaum D, Mirouse A, Fieschi C,

Malphettes M, Vercellino L, Meignin V, Gerard L, Galicier L. The full spectrum

of Castleman disease: 273 patients studied over 20 years. Br J Haematol

2018;180:206-216.

23. Fajgenbaum DC, Uldrick TS, Bagg A, Frank D, Wu D, Srkalovic G, Simpson D,

Liu AY, Menke D, Chandrakasan S, Lechowicz MJ, Wong RSM, Pierson

S, Paessler M, Rossi JF, Ide M, Ruth J, Croglio M, Suarez A, Krymskaya

V, Chadburn A, Colleoni G, Nasta S, Jayanthan R, Nabel CS, Casper C,

Dispenzieri A, Fossa A, Kelleher D, Kurzrock R, Voorhees P, Dogan A, Yoshizaki

K, van Rhee F, Oksenhendler E, Jaffe ES, Elenitoba-Johnson KSJ, Lim MS.

International, evidence-based consensus diagnostic criteria for HHV-8–

negative/idiopathic multicentric Castleman disease. Blood 2017;129:1646-

1657.

135


PERSPECTIVE

DOI: 10.4274/tjh.galenos.2021.2021.0683

Turk J Hematol 2022;39:136-139

Gaucher Disease for Hematologists

Hematologlar için Gaucher Hastalığı

Gül Nihal Özdemir 1 , Eren Gündüz 2

1İstinye University Faculty of Medicine, Department of Pediatric Hematology Oncology, İstanbul, Turkey

2Eskisehir Osmangazi University Faculty of Medicine, Department of Hematology, Eskişehir, Turkey

Abstract

Gaucher disease (GD) is a rare hereditary lysosomal storage disease

that arises due to deficiency of glucocerebrosidase. Early diagnosis

is very important for starting proper treatment and preventing

complications. Splenomegaly, anemia, and thrombocytopenia are the

most common findings in GD and so most patients are initially referred

to hematologists. The Turkish Society of Hematology established its

Rare Hematological Diseases Subcommittee in 2015. One of the main

topics of this subcommittee was to increase and improve awareness

and education of rare diseases among hematologists in Turkey.

This review presents GD with an overview of its clinical features,

pathophysiology, and treatment options for hematologists.

Keywords: Gaucher disease, Anemia, Thrombocytopenia, Splenomegaly

Öz

Gaucher hastalığı (GH) nadir görülen kalıtsal bir lizozomal depo

hastalığıdır. Gaucher hastaları sıklıkla geç tanı alırlar. Erken teşhis;

uygun tedaviye başlamak, komplikasyonları ve hastalığın ilerlemesini

önlemek için önemlidir. Splenomegali, anemi ve trombositopeni

GH’de en sık görülen bulgulardır, bu nedenle çoğu hasta başlangıçta

hematologlara sevk edilir. Türk Hematoloji Derneği 2015 yılında “Nadir

hematolojik hastalıklar alt komitesini” kurmuştur. Alt komitenin ana

amaçlarında biri Türkiye’deki hematologların nadir görülen hastalıklar

konusunda bilinçlendirilmesi ve eğitimlerinin artırılması olmuştur. Bu

derleme, hematologlar için GH’nin klinik özellikleri, patofizyolojisi ve

tedavi seçeneklerine bir bakış sunmaktadır.

Anahtar Sözcükler: Gaucher hastalığı, Anemi, Trombositopeni,

Splenomegali

Introduction

Gaucher disease (GD) is a rare hereditary lysosomal storage

disease [1]. It occurs due to deficiency of the lysosomal enzyme

glucocerebrosidase and the results of several compounds

related to the substrate accumulating in cell lysosomes. The

glucocerebrosidase 1 (GBA1) gene encoding glucocerebrosidase

is located on chromosome 1. This disease has traditionally

been classified into subtypes according to clinical findings,

course, and the patient’s ethnic origin. Type 1 GD (GD1, adult

type) is the most prevalent form. Visceral findings such as

hepatosplenomegaly, cytopenia, and bone disease are observed.

While cases of GD1 lack involvement of the central nervous

system, recent studies have shown that Parkinson disease and

peripheral neuropathy are more common in these cases [2]. Type

2 GD (GD2, infantile) is the most severe form and begins within

the first 6 months of life with a life expectancy of <2 years [3].

In addition to enlargement of the spleen and liver, progressive

neurological findings are observed. In type 3 GD (GD3, juvenile),

patients have both visceral and neurological findings with

longer survival. Recently, however, this classification according

to subtypes is being abandoned. The three subtypes are now

thought to be continuations of each other within the spectrum

of the same disease, rather than disorders with different

phenotypes.

Patients with GD often have delays in diagnosis of up to 10

years [4]. Early diagnosis is important for starting proper

treatment and preventing complications as well as disease

progression. Splenomegaly, anemia, and thrombocytopenia are

the most common findings in GD so most patients are initially

referred to hematologists with a differential diagnosis of

leukemia, lymphoma, or immune thrombocytopenia [5,6]. The

Turkish Society of Hematology established a Rare Hematological

Diseases Subcommittee in 2015. One of the main tasks of the

subcommittee was to increase and improve awareness and

education of rare diseases among hematologists in Turkey and

GD was selected as one of the target diseases within this project.

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Gül Nihal Özdemir, M.D., İstinye University Faculty of Medicine,

Department of Pediatric Hematology Oncology, İstanbul, Turkey

E-mail : gul.ozdemir@istinye.edu.tr ORCID: orcid.org/0000-0002-3204-4353

Received/Geliş tarihi: February 6, 2022

Accepted/Kabul tarihi: April 18, 2022

136


Turk J Hematol 2022;39:136-139

Özdemir G.N. and Gündüz E.: Gaucher Disease

Weekend courses, online educational meetings, and guidelines

for the diagnosis and treatment of GD were organized. This

review presents GD with an overview of its clinical features,

pathophysiology, and treatment options for hematologists.

Clinical Findings

There is a distinct phenotypic diversity in GD that cannot be

fully explained by the genotype [7,8,9]. The clinical findings

are shown in Table 1 [7]. The visceral organs, bone marrow

(Figure 1), and bones are affected in almost all cases.

The most common finding is splenomegaly [10]. Isolated

thrombocytopenia alone is the most common cytopenia.

Anemia and rarely leukopenia may be seen. Bone findings

may present as diffuse bone pain and pain attacks associated

with osteonecrosis. Osteolytic lesions, pathological fractures,

and compression fractures are more common in patients who

have undergone splenectomy. Many patients have growth

retardation and delayed puberty [11]. Interstitial lung disease

may be detected in rare cases [12].

Diagnosis

Laboratory and radiological findings are summarized in

Table 2 and Table 3. Serum angiotensin-converting enzyme

and particularly its tartrate-resistant isoenzymes may

be increased [13]. Levels of chitotriosidase, an enzyme

secreted from lipid-laden macrophages, are elevated [14].

Hyperferritinemia is frequently encountered in GD [15].

Lyso-GL1 (glucosylsphingosine), which is a downstream metabolic

product of glucosylceramide, has been identified as a promising

biomarker for the diagnosis and monitoring of patients with GD

in recent years [16]. The definitive diagnosis of GD is made with

glucocerebrosidase enzyme detection and genetic mutation

analysis. Glucocerebrosidase can be examined in peripheral

leukocytes or skin fibroblasts. For this, it is necessary to take a

dry blood sample on filter papers [17]. Genetic analysis provides

additional confirmation of the diagnosis and is also helpful for

genetic counseling and the detection of carriers [18]. GBA1 is

the only gene known to mutate in GD and the most common

Table 1. Signs and symptoms of Gaucher disease.

Splenomegaly 85%

Hepatomegaly 63%

Thrombocytopenia 68%

Anemia 34%

Bleeding

Osteopenia 55%

Bone pain 33%

Pathological fractures 7%

Bone crises 7%

Growth retardation 36%

Frequent (no percentage reported)

mutation is the N370S mutation [7]. Lipid-loaded macrophages

can be seen in the bone marrow in GD, but that is not a specific

finding. Pseudo-Gaucher cells can be seen in other diseases [20].

Bone marrow aspiration/biopsy is not required for the definitive

diagnosis but may be performed to rule out other diseases.

Treatment

The goals of treatment are the elimination of symptoms,

prevention of complications, and improvement of quality

of life [21]. Due to the heterogeneity of the disease and the

uncertainty of disease progression, the management should

be individualized. Enzyme replacement therapy ameliorates

most of the manifestations of GD1 and improves quality of

life [22]. Treatment is not recommended for GD2 patients as it

does not stop the clinical course. Enzyme replacement therapy

may be beneficial for GD3 patients who have chronic visceral

manifestations. Indications for starting treatment in cases of

GD1 are considered according to the severity of the disease

at the initial evaluation or according to the progression of

Table 2. Laboratory findings in cases of Gaucher disease.

• Cytopenia

* Anemia

* Thrombocytopenia

* Leukopenia

* Bicytopenia/pancytopenia

• Coagulation disorders

• Elevated liver enzymes

• Increase in serum ACE level (especially tartrate-resistant

isoenzymes)

• Increase in acid phosphatase activity

• Hyperferritinemia

• Increase in chitotriosidase

• Poly- and monoclonal gammopathy

• Lipid-loaded macrophages in tissues (bone marrow, liver, spleen)

Table 3. Radiological findings in cases of Gaucher disease.

• Bone radiography

° Erlenmeyer flask deformity

° Bone fractures and lytic lesions

• Bone magnetic resonance imaging (MRI)

° Bone marrow involvement

° Bone infarcts

° Osteonecrosis

• Dual-energy X-ray absorptiometry (DEXA)

° Osteopenia

• Abdominal ultrasonography (USG)

° Hepatomegaly

° Splenomegaly

• Echocardiography

° Pulmonary hypertension

• Chest X-ray/thorax computed tomography (CT)

° Lung involvement

137


Özdemir G.N. and Gündüz E.: Gaucher Disease

Turk J Hematol 2022;39:136-139

the disease. The severity of the disease can be evaluated with

scoring systems [23]. Enzyme replacement therapy for GD1

may include imiglucerase, velaglucerase alfa, and taliglucerase

alfa [24,25,26]. There is no consensus on the optimal dose or

frequency in the administration of recombinant enzymes.

The recommended dose for imiglucerase is 15-60 units of

enzyme/kg every 2 weeks intravenously. The ideal dose has

been set at 60 units/kg in most studies, but good response

was also shown at lower doses [27]. Treatment is life-long and

interruptions are not recommended. A small percentage of

patients may develop antibodies (15%) [28]. Glucosylceramide

synthase inhibitors (miglustat and eliglustat) are used for

substrate reduction therapy [29,30]. They reduce the amount of

substrate and prevent the symptoms that develop accordingly.

Miglustat is approved for patients with mild to moderate GD1

who cannot undergo enzyme therapy and also for the small

group of patients for whom enzyme therapy is unsuitable due

to adverse events or venous access problems [31]. The role of

splenectomy has decreased with the availability of enzyme

replacement therapy. Some studies have shown that total

splenectomy worsens bone findings in GD [32]. Bone marrow

transplantation offer the potential of cure, but no clinical trials

to date have assessed its safety and efficacy in comparison to

enzyme replacement therapy or substrate reduction therapy

[33].

Conclusion

Gaucher disease is a rare but treatable metabolic disease. High

levels of suspicion are necessary for early diagnosis as this disease

may present with different clinical findings. Early treatment will

be beneficial in preventing irreversible complications.

Authorship Contributions

Concept: G.N.Ö.; Design: G.N.Ö., E.G.; Literature Search: E.G.;

Writing: G.N.Ö.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Grabowski GA. Gaucher disease: gene frequencies and genotype/phenotype

correlations. Genet Test 1997;1:5-12.

2. Biegstraaten M, van Schaik IN, Aerts JM, Hollak CE. ‘Non-neuronopathic’

Gaucher disease reconsidered. Prevalence of neurological manifestations in

a Dutch cohort of type I Gaucher disease patients and a systematic review

of the literature. J Inherit Metab Dis 2008;31:337-349.

3. Sidransky E. New perspectives in type 2 Gaucher disease. Adv Pediatr

1997;44:73-107.

4. Mistry PK, Sadan S, Yang R, Yee J, Yang M. Consequences of diagnostic

delays in type 1 Gaucher disease: the need for greater awareness among

hematologists-oncologists and an opportunity for early diagnosis and

intervention. Am J Hematol 2007;82:697-701.

5. Thomas AS, Mehta A, Hughes DA. Gaucher disease: haematological

presentations and complications. Br J Haematol 2014;165:427-440.

Figure 1. Bone marrow aspirate showing a number of large

macrophages laden with cerebrosides (arrows: Gaucher cells) in a

patient with Gaucher disease.

6. Linari S, Castaman G. Hematological manifestations and complications of

Gaucher disease. Expert Rev Hematol 2016;9:51-58.

7. Charrow J, Andersson HC, Kaplan P, Kolodny EH, Mistry P, Pastores G,

Rosenbloom BE, Scott CR, Wappner RS, Weinreb NJ, Zimran A. The Gaucher

138


Turk J Hematol 2022;39:136-139

Özdemir G.N. and Gündüz E.: Gaucher Disease

Registry: Demographics and disease characteristics of 1698 patients with

Gaucher disease. Arch Intern Med 2000;160:2835-2843.

8. Alfonso P, Aznarez S, Giralt M, Pocovi M, Giraldo P; Spanish Gaucher’s

Disease Registry. Mutation analysis and genotype/phenotype relationships

of Gaucher disease patients in Spain. J Hum Genet 2007;52:391-396.

9. Nalysnyk L, Rotella P, Simeone JC, Hamed A, Weinreb N. Gaucher disease

epidemiology and natural history: a comprehensive review of the literature.

Hematology 2017;22:65-73.

10. Motta I, Filocamo M, Poggiali E, Stroppiano M, Dragani A, Consonni

D, Barcellini W, Gaidano G, Facchini L, Specchia G, Cappellini MD;

Splenomegaly Gaucher Disease Study Group. A multicentre observational

study for early diagnosis of Gaucher disease in patients with splenomegaly

and/or thrombocytopenia. Eur J Haematol 2016;96:352-359.

11. Baris HN, Cohen IJ, Mistry PK. Gaucher disease: the metabolic defect,

pathophysiology, phenotypes and natural history. Pediatr Endocrinol Rev

2014;12(Suppl 1):72-81.

12. Miller A, Brown LK, Pastores GM, Desnick RJ. Pulmonary involvement in

type 1 Gaucher disease: functional and exercise findings in patients with

and without clinical interstitial lung disease. Clin Genet 2003;63:368-376.

13. Danilov SM, Tikhomirova VE, Metzger R, Naperova IA, Bukina TM, Goker-

Alpan O, Tayebi N, Gayfullin NM, Schwartz DE, Samokhodskaya LM, Kost

OA, Sidransky E. ACE phenotyping in Gaucher disease. Mol Genet Metab

2018;123:501-510.

14. van Dussen L, Hendriks EJ, Groener JE, Boot RG, Hollak CE, Aerts JM. Value

of plasma chitotriosidase to assess non-neuronopathic Gaucher disease

severity and progression in the era of enzyme replacement therapy. J Inherit

Metab Dis 2014;376:991-1001.

15. Regenboog M, van Kuilenburg AB, Verheij J, Swinkels DW, Hollak CE.

Hyperferritinemia and iron metabolism in Gaucher disease: potential

pathophysiological implications. Blood Rev 2016;30:431-437.

16. Elstein D, Mellgard B, Dinh Q, Lan L, Qiu Y, Cozma C, Eichler S, Böttcher

T, Zimran A. Reductions in glucosylsphingosine (lyso-Gb1) in treatmentnaïve

and previously treated patients receiving velaglucerase alfa for type

1 Gaucher disease: data from phase 3 clinical trials. Mol Genet Metab

2017;122:113-120.

17. Herrera D, Monaga M, Campos D, Pampín Y, González EC, Lavaut K.

Ultramicro-fluorometric assay for the diagnosis of Gaucher disease in dried

blood spots on filter paper. J Neonatal Perinatal Med 2013;6:61-67.

18. Yoshida S, Kido J, Matsumoto S, Momosaki K, Mitsubuchi H, Shimazu T,

Sugawara K, Endo F, Nakamura K. Prenatal diagnosis of Gaucher disease

using next-generation sequencing. Pediatr Int 2016;58:946-949.

19. Dahl N, Lagerström M, Erikson A, Pettersson U. Gaucher disease type III

(Norrbottnian type) is caused by a single mutation in exon 10 of the

glucocerebrosidase gene. Am J Hum Genet 1990;47:275-278.

20. Gören Şahin D, Üsküdar Teke H, Karagülle M, Andıç N, Gündüz E, Işıksoy

S, Balić M, Akay OM. Gaucher cells or pseudo-Gaucher cells: that’s the

question. Turk J Hematol 2014;31:428-429.

21. Pastores GM, Weinreb NJ, Aerts H, Andria G, Cox TM, Giralt M, Grabowski

GA, Mistry PK, Tylki-Szymańska A. Therapeutic goals in the treatment of

Gaucher disease. Semin Hematol 2004;41(4 Suppl 5):4-14.

22. Charrow J, Scott CR. Long-term treatment outcomes in Gaucher disease.

Am J Hematol 2015;90(Suppl 1):S19-24.

23. Di Rocco M, Giona F, Carubbi F, Linari S, Minichilli F, Brady RO, Mariani

G, Cappellini MD. A new severity score index for phenotypic classification

and evaluation of responses to treatment in type I Gaucher disease.

Haematologica 2008;93:1211-1218.

24. Serratrice C, Carballo S, Serratrice J, Stirnemann J. Imiglucerase in the

management of Gaucher disease type 1: an evidence-based review of its

place in therapy. Core Evid 2016;11:37-47.

25. Smith L, Rhead W, Charrow J, Shankar SP, Bavdekar A, Longo N, Mardach

R, Harmatz P, Hangartner T, Lee HM, Crombez E, Pastores GM. Long-term

velaglucerase alfa treatment in children with Gaucher disease type 1 naïve

to enzyme replacement therapy or previously treated with imiglucerase.

Mol Genet Metab 2016;117:164-171.

26. Zimran A, Elstein D. Management of Gaucher disease: enzyme replacement

therapy. Pediatr Endocrinol Rev 2014;12(Suppl 1):82-87.

27. de Fost M, Hollak CE, Groener JE, Aerts JM, Maas M, Poll LW, Wiersma MG,

Häussinger D, Brett S, Brill N, vom Dahl S. Superior effects of high-dose

enzyme replacement therapy in type 1 Gaucher disease on bone marrow

involvement and chitotriosidase levels: a 2-center retrospective analysis.

Blood 2006;108:830-835.

28. Starzyk K, Richards S, Yee J, Smith SE, Kingma W. The long-term international

safety experience of imiglucerase therapy for Gaucher disease. Mol Genet

Metab 2007;90:157-163.

29. Van Rossum A, Holsopple M. Enzyme replacement or substrate reduction?

A review of Gaucher disease treatment options. Hosp Pharm 2016;51:553-

563.

30. Shemesh E, Deroma L, Bembi B, Deegan P, Hollak C, Weinreb NJ, Cox TM.

Enzyme replacement and substrate reduction therapy for Gaucher disease.

Cochrane Database Syst Rev 2015;2015:CD010324.

31. Heitner R, Elstein D, Aerts J, van Weely S, Zimran A. Low-dose

N-butyldeoxynojirimycin (OGT 918) for type I Gaucher disease. Blood Cells

Mol Dis 2002;28:127-133.

32. Fleshner PR, Aufses AH Jr, Grabowski GA, Elias R. A 27-year experience with

splenectomy for Gaucher’s disease. Am J Surg 1991;161:69-75.

33. Somaraju UR, Tadepalli K. Hematopoietic stem cell transplantation for

Gaucher disease. Cochrane Database Syst Rev 2017;10:CD006974.

139


IMAGES IN HEMATOLOGY

DOI: 10.4274/tjh.galenos.2020.2020.0693

Turk J Hematol 2022;39:140-141

Gallbladder Involvement of Diffuse Large B-Cell Lymphoma with

18

F-FDG PET/CT

18

F-FDG PET/CT Görüntüleme ile Diffüz Büyük B Hücreli Lenfomanın Safra Kesesi Tutulumu

Esra Arslan, Göksel Alçin, Tamer Aksoy, Tevfik Fikret Çermik

University of Health Sciences Turkey, İstanbul Training and Research Hospital, Clinic of Nuclear Medicine, İstanbul, Turkey

Figure 1. Involvement of multiple lymph nodes with high

radiopharmaceutical uptake in supra- and infradiaphragmatic

lymphatic stations.

CT: Computed tomography, PET: positron emission tomography, F: fusion,

MIP: maximum intensity projection.

Figure 2. Increased 18 F-FDG uptake was observed in newly

developed metastatic nodes in addition to previous lesions in

the supra/infradiaphragmatic regions on posttreatment PET/CT.

Additionally, focal 18 F-FDG uptake (dashed arrows) in the upper

outer quadrant of the right breast and unexpected gallbladder

(GB) wall involvement (arrows) were observed.

CT: Computed tomography, PET: positron emission tomography, F: fusion,

MIP: maximum intensity projection.

18

F-Fluoro-2-deoxy-glucose positron emission tomography/

computed tomography ( 18 F-FDG PET/CT) is widely used in staging,

restaging, and evaluation of treatment response in patients with

lymphoma. In this case report, a 59-year-old woman diagnosed

with diffuse large B-cell lymphoma (DLBCL) with gallbladder

involvement is presented. Immunohistochemical analysis

showed strongly positive CD79a, bcl-6, MUM1, bcl-2, and CD43;

weakly positive CD20 and CD5 staining; negative c-myc; and

Ki-67 of 85%. Interim 18 F-FDG PET/CT showed involvement of

multiple lymph nodes with high radiopharmaceutical uptake in

supra- and infradiaphragmatic lymphatic stations (Figure 1). On

the other hand, increased 18 F-FDG uptake was observed in newly

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Esra Arslan, M.D., University of Health Sciences Turkey, İstanbul

Training and Research Hospital, Clinic of Nuclear Medicine, İstanbul, Turkey

E-mail : dresraarslan@gmail.com ORCID: orcid.org/0000-0002-9222-8883

Received/Geliş tarihi: November 19, 2020

Accepted/Kabul tarihi: December 28, 2020

140


Turk J Hematol 2022;39:140-141

Arslan E. et al: Gallbladder Involvement of Diffuse Large B-Cell Lymphoma

developed metastatic nodes in addition to previous lesions in

the supra/infradiaphragmatic regions on posttreatment PET/CT.

Additionally, focal 18 F-FDG uptake (dashed arrows) in the upper

outer quadrant of the right breast and unexpected gallbladder

(GB) wall involvement (arrows) were observed (Figure 2).

18

F-FDG PET/CT is a diagnostic method for detecting metastatic

lesions in GB [1,2,3]. Although the involvement of the breasts

and thyroid in non-Hodgkin’s lymphoma is frequently reported,

involvement of the GB is extremely rare [4,5,6]. Al-Katib

et al. [7] detected extranodal involvement of the GB in an

83-year-old male with intravascular large B-cell lymphoma. Bai

et al. [8] reported increased 18 F-FDG uptake in the GB without

luminal pathology in a 15-year-old girl with Hodgkin’s disease.

In our case, 18 F-FDG PET/CT imaging revealed DLBCL-related

involvement in the GB. PET/CT is a useful tool for demonstrating

unexpected organ involvements, such as in the GB.

18

Keywords: F-FDG PET/CT, Gallbladder,

lymphoma

Non-Hodgkin’s

Anahtar Sözcükler: 18 F-FDG PET/BT, Safra kesesi, Non-Hodgkin

lenfoma

Ethics

Informed Consent: The patient provided verbal and written

consent for the use of the medical findings for research purposes.

Authorship Contributions

Surgical and Medical Practices: T.A.; Concept: E.A.,

T.F.Ç., Design: E.A.; Data Collection or Processing: G.A.;

Analysis or Interpretation: T.A., T.F.Ç.; Literature Search: G.A.;

Writing: E.A.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Chung PH, Srinivasan R, Linehan WM, Pinto PA, Bratslavsky G. Renal cell

carcinoma with metastases to the gallbladder: four cases from the National

Cancer Institute (NCI) and review of the literature. Urol Oncol 2012;30:476-

481.

2. Shaikh F, Awan O, Khan SA. 18F-FDG PET/CT imaging of gallbladder

adenocarcinoma-a pictorial review. Cureus 2015;7:e298.

3. Kursat O, Engin A, Nuri A, Seref K, Erkan O. Watch out for the unexpected:

sole gallbladder metastasis in a patient with malignant melanoma striked

by FDG-PET. J Nucl Med Radiat Ther 2015;6:2.

4. Dravid NV, Ningurkar NU, Nikumbh DB, Gadre AS. Extranodal primary non

hodgkin lymphoma of breast: multimodal approach to diagnosis. Indian J

Pathol Oncol 2018;5:349-351.

5. Binesh F, Akhavan A, Navabii H. Extranodal marginal zone B cell lymphoma

of MALT type with extensive plasma cell differentiation in a man with

Hashimoto’s thyroiditis. BMJ Case Rep 2011;2011:bcr0520114277.

6. Pezzuto R, Di Mauro D, Bonomo L, Patel A, Ricciardi E, Attanasio A, Manzelli

A. An unusual case of primary extranodal lymphoma of the gallbladder.

Hematol Rep 2017;9:6972

7. Al-Katib S, Colvin R, Sokhandon F. Intravascular large B-cell lymphoma

presenting with diffuse gallbladder wall thickening: a case report and

literature review. Case Rep Radiol 2018;2018:2494207.

8. Bai X, Wang X, Zhuang H. FDG accumulation in the lumen of the gallbladder

without related pathology. Clin Nucl Med 2018;43:383-385.

141


IMAGES IN HEMATOLOGY

DOI: 10.4274/tjh.galenos.2022.2021.0530

Turk J Hematol 2022;39:142-143

Mast Cell Leukemia in a Patient with Teratoma

Teratomlu Bir Hastada Mast Hücreli Lösemi

Yan Shen*, Zhenni Wang*, Huijun Lin*, Jinlin Liu

Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou

Medical College), Hangzhou, China

*These authors contributed equally to this work.

Figure 1. Blood smear showed peripheral blood involvement (40%) by promyelocyte-like leukemia cells characterized by a spindle shape

with packed and polar granule aggregates (A), dandelion-like circulating round cells (B), and prominent vacuoles in the cytoplasm (C).

Bone marrow aspirate smear showed clusters of cells resembling metastatic cells (30%) (D, E). Toluidine blue staining of the bone marrow

revealed the cytoplasm to be chemoreactive for mast cells (F).

A 29-year-old man was admitted to the hospital with the

complaints of left upper chest pain, asthenia, melena, and red

but not itchy rash across the whole body. Initial examination

for thrombocytopenia (38x10 9 /L), normocytic anemia (8.2 g/dL),

and white blood cell count (15.6x10 9 /L) yielded a diagnosis

of acute promyelocyte leukemia (APL). Physical examination

revealed hepatosplenomegaly.

A blood smear showed peripheral blood involvement (40%) by

promyelocyte-like leukemia cells (Figures 1A-1C). These cells

were characterized by a spindle shape with packed and polar

granule aggregates (Figure 1A), dandelion-like circulating round

cells (Figure 1B), and prominent vacuoles in the cytoplasm

(Figure 1C). A bone marrow aspirate smear showed clusters

of cells resembling metastatic cells (30%) (Figures 1D and 1E).

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Jinlin Liu, M.D., Ph.D., Zhejiang Provincial People’s Hospital,

Hangzhou Medical College, Cytology Unit, Department of Clinical Laboratory, Hangzhou, China

Phone : 0086-18768137609

E-mail : liujinlinhz@163.com ORCID: orcid.org/0000-0003-0502-4813

Received/Geliş tarihi: September 15, 2021

Accepted/Kabul tarihi: February 22, 2022

142


Turk J Hematol 2022;39:142-143

Shen Y. et al: Mast Cell Leukemia in a Patient with Teratoma

Additionally, chromosome analysis revealed a normal karyotype.

Considering that the patient had a history of malignant

mediastinal teratoma, teratoma metastasis to the bone marrow

was suspected. However, the flow cytometry for these abnormal

cells was positive for CD117, CD33, CD13, CD11c, CD4, CD38,

CD22, CD25, and CD2. Furthermore, toluidine blue staining of

the bone marrow revealed the cytoplasm to be chemoreactive

for mast cells (Figure 1F). Together with the KIT D816V mutation

identified in this patient, the diagnosis of mast cell leukemia

(MCL) was made according to the 2016 systemic mastocytosis

criteria of the World Health Organization. Thus, this patient was

treated with dasatinib, lucitanib, and dexamethasone, and later

with midostaurin, but no improvement was achieved and he

died two months later.

MCL is an aggressive form of systemic mastocytosis. Increasing

promyelocyte-like blasts in the peripheral blood were initially

suggestive of APL, and clusters of blast cells in the bone marrow

were suggestive of the metastasis of teratoma, factors that

could lead to the incorrect diagnosis of MCL. However, as

atypical features were of relevance for other hematological

malignancies, toluidine blue staining, immunostaining, and

KIT D816V mutation were evaluated to further support the

diagnosis of MCL.

Keywords: Mast cell leukemia, Teratoma

Anahtar Sözcükler: Mast hücreli lösemi, Teratom

Acknowledgments

The authors appreciate the support of the members of the

Department of Clinical Laboratory, Zhejiang Provincial People’s

Hospital.

Ethics

Informed Consent: This study did not involve personal

information; only laboratory data were reported. Patient

consent was therefore waived, and this waiver was approved by

the Ethics Committee of Zhejiang Provincial People’s Hospital.

Authorship Contributions

Data Collection or Processing: Y.S., Z.W., H.L.; Writing: J.L.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: This work was supported by the General

Research Program of the Zhejiang Provincial Department of

Health (no. 2018KY249).

143


LETTERS TO THE EDITOR

Turk J Hematol 2022;39:144-151

Caution Regarding the Difference Between Flower-Like

Lymphocytes and Flower-Like Plasma Cells

Çiçek Gibi Lenfositler ile Çiçek Gibi Plazma Hücreleri Arasındaki Farka Dikkat!

Jingnan Zhu 1,2 , Zhang Li 3 , Yong Wang 2 , Jinlin Liu 4

1Zhejiang Chinese Medical University, College of Medical Technology, Hangzhou, China

2Shangyu People’s Hospital of Shaoxing, Department of Clinical Laboratory, Shaoxing, China

3People’s Hospital of Zhenhai, Department of Clinical Laboratory, Ningbo, China

4Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Department of Clinical Laboratory, Hangzhou, China

To the Editor,

We attentively read the paper by Sall et al. [1] recently

published in Turkish Journal of Hematology. These authors

reported a case of multiple myeloma showing abnormal

plasma cells with flower-shaped nuclear features. These cells

with flower-shaped nuclei have also been reported in cases

of plasma cell leukemia in previous studies [2,3,4,5]. The

authors described the morphological features of flower-like

cells in order to help pathologists identify them for diagnostic

purposes. However, the flower-like plasma cells with condensed

chromatin presented in Figure 1B of the original study [1]

look like flower-like lymphocytes. Furthermore, these cells

had not been confirmed by immunocytochemical staining.

CD20 or CD138 immunocytochemistry could have been used

to confirm whether these cells were flower-like plasma cells or

flower-like lymphocytes, as previously reported [6]. Therefore,

the differential diagnosis between flower-like lymphocytes and

flower-like plasma cells should be approached with caution.

As we know, the presence of flower-like lymphocytes is a wellknown

morphological feature in the peripheral blood of patients

with adult T-cell leukemia/lymphoma (ATLL) [7]. However,

evaluation of flower-like lymphocytes must be performed

carefully because these cells appear not only in ATLL patients but

also in patients with various other diseases entailing reactive or

neoplastic lesions, or even in healthy individuals [6,8].

In our experience, these flower-like lymphocytes are frequently

found in the peripheral blood in routine daily blood examination.

We can consider the daily peripheral blood of a hepatitis B

and an anemia patient for example. Flower-like lymphocytes

are clearly visible in the peripheral blood (Figures 1A and 1B).

Figure 1. Peripheral blood smears of a hepatitis B patient (A) and an anemia patient (B) reveal flower-like lymphocytes. These flower-like

lymphocytes clearly have flower-like nuclei (Wright-Giemsa staining, 1000 x ).

144


Turk J Hematol 2022;39:144-151

LETTERS TO THE EDITOR

Therefore, when encountering these flower-like cells in blood

smears, flower-like lymphocytes should also be considered for

the differential diagnosis of flower-like plasma cells.

Keywords: Flower-like lymphocyte, Flower-like plasma cell,

Difference

Anahtar Sözcükler: Çiçek benzeri lenfosit, Çiçek benzeri plazma

hücresi, Fark

Acknowledgment

The authors appreciate the encouragement and support of

the members of the cytology unit of the Clinical Laboratory

Department, Zhejiang Provincial People’s Hospital.

Ethics

Authorship Contributions

Surgical and Medical Practices: J.Z., Concept: J.L.; Design: Y.W.;

Data Collection or Processing: J.Z.; Analysis or Interpretation:

J.Z.; Literature Search: Z.L.; Writing: J.L.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Sall A, Seck M, Samb D, Faye B, Gadji M, Diop S, Touré AO. Flower-like

plasma cell nuclei in multiple myeloma. Turk J Hematol 2021;38:153-154.

2. Gajendra S. Flower-like plasma cell: a comment. Turk J Hematol

2021;38:228-229.

3. De Miguel Sánchez C, Robles de Castro D, Pisón Herrero C, Pérez Persona

E, Salcedo Cuesta L, Guinea de Castro JM. Primary plasma cell leukaemia

presenting with flower-shaped nuclei. Br J Haematol 2021;193:689.

4. Shibusawa M. Plasma cell leukaemia presenting as flower-shaped plasma

cells mimicking adult T-cell leukaemia or lymphoma. Lancet Haematol

2020;7:e270.

5. Delhommeau F, Huguet S, Gachet J, van den Akker J, Lagrange M. Primary

plasma cell leukemia mimicking an adult T-cell leukemia-lymphoma: a case

report. Acta Cytol 2010;54:187-189.

6. Kobayashi M, Horikawa M, Uehara T, Honda T, Kobayashi T, Sakai H. Flowerlike/clover

leaf lymphocytes appear in various diseases: cerebrospinal fluid

cytology case with review of the literature. Diagn Cytopathol 2015;43:88-

90.

7. Dahmoush L, Hijazi Y, Barnes E, Stetler-Stevenson M, Abati A. Adult T-cell

leukemia/lymphoma: a cytopathologic, immunocytochemical, and flow

cytometric study. Cancer 2002;96:110-116.

8. Yaghmour G, Thind R, Kryvenko ON, Ayyad H, Chen A, Kuriakose P. All that

glitters is not gold: differential diagnosis of clover-leaf lymphocytes. Int J

Hematol 2013;97:679-680.

To the Editor,

We thank Zhu et al. for their interest in our article.

We agree with their observations regarding lymphocytes.

However, in our patient, these flower-like plasma cells were

found only in the marrow. Cytochemistry was not performed,

but all relevant cytological evidence was visible on the smear:

for example, rouleaux, plasma cells, and nuclear budding. Upon

immunophenotyping, a homogeneous population of tumor

plasma cells was found. HBV and HTLV1 serologies were also

evaluated and were negative.

In the absence of clinical signs such as bone pain and

spontaneous fractures and in light of the monoclonal gammaglobulin

peak, cytologic immunophenotypic evidence, and

serological negativity, we agree that flower-like lymphocytes

could be considered in the differential diagnosis.

Sincerely,

Abibatou Sall, Moussa Seck, Diama Samb, Blaise Faye, Macoura

Gadji, Saliou Diop, Awa Oumar Touré

©Copyright 2021 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Yong Wang, M.D., Shangyu People’s Hospital of Shaoxing,

Department of Clinical Laboratory, Shaoxing, China / Jinlin Liu, M.D., Ph.D., Zhejiang Provincial People’s

Hospital, Hangzhou Medical College, Department of Clinical Laboratory, Hangzhou, China

E-mail : 291615783@qq.com/liujinlinhz@163.com ORCID: orcid.org/0000-0001-8530-3809/

Received/Geliş tarihi: March 2, 2022

Accepted/Kabul tarihi: March 17, 2022

DOI: 10.4274/tjh.galenos.2022.2022.0087

145


LETTERS TO THE EDITOR

Turk J Hematol 2022;39:144-151

Does Treatment of Hepatitis C Reduce Inhibitor Titers in

Hemophilia A?

Hepatit C Tedavisi Hemofili A’da İnhibitör Titrelerini Azaltır mı?

Memiş Hilmi Atay 1 , Emine Türkoğlu 2 , Engin Kelkitli 1

1Ondokuz Mayıs University Faculty of Medicine, Department of Internal Medicine, Division of Hematology, Samsun, Turkey

2Tokat Gaziosmanpaşa University Faculty of Medicine, Department of Infectious Diseases, Tokat, Turkey

To the Editor,

Hemophilia A is an X-linked recessive disorder characterized by

a deficiency of factor VIII in plasma [1]. It has been reported

that approximately 30% of patients who receive recombinant or

plasma-derived factor VIII prophylaxis may develop neutralizing

antibodies or inhibitors [2]. Bleeding episodes in patients with

low-responding inhibitors are treated with factor VIII, whereas

bleeding episodes in those with high-responding inhibitors are

treated with bypassing agents, such as recombinant activated

factor VIIa (rFVIIa) [3]. Here we discuss the case of a patient

with factor VIII inhibitors whose need for factor replacement

decreased with the treatment of chronic hepatitis C.

A 47-year-old male patient underwent surgery due to a fracture

of the right elbow. He had factor VIII deficiency and was first

diagnosed at the age of 10 with a factor VIII level of 4 IU. He

was treated with factor replacement due to bleeding episodes.

He did not have major bleeding episodes or major surgery.

The preoperative factor FVIII level was reported as 1.2 IU/mL.

Inhibitors could not be studied. Before the surgery, he was

given factor VIII concentrate loading of 3000 units and 2x1500

units (50 U/kg) for one week. On the seventh postoperative

day, the levels of factor VIII and inhibitor were assessed due to

ongoing bleeding from the operation site. Factor VIII was found

to be 2 IU/mL and the inhibitor to be 19.2 BU/mL. No factor

VIII inhibitor had been detected in the patient previously. The

treatment was switched to a bypassing agent, rFVIIa, at 5400 µg

(90 µg/kg/dose) with 3 doses to be administered every 3 hours.

Once the bleeding was under control, the rFVIIa treatment was

administered with decreasing frequency. The patient was HCVpositive

with HCV RNA of 550,000 units of copies. He tested

negative for hepatitis B surface antigens and HIV antibodies. The

patient, who had HCV genotype 2, was started on direct-acting

antiviral (DAA) drug treatment with glecaprevir/pibrentasvir at

1x3 tablets daily. After starting DAA treatment, the patient’s

need for factor rFVIIa decreased (Figure 1). As a result, the dose

frequency was decreased and treatment with rFVIIa continued

twice a week. After 8 weeks, HCV RNA was not detected. The

level of factor VIII inhibitor was detected as 1.2 BU/mL.

It has been found that patients who are HCV-positive with factor

VIII inhibitors have a complex immune profile characterized

by higher levels of pro-inflammatory and anti-inflammatory

cytokines compared to those who are not [4,5]. The cure rate of

chronic hepatitis C has reached 90% with DAA therapy [6]. Triple

treatment with interferon, telaprevir, and ribavirin has been

reported to result in a decrease in the level of inhibitors and the

need for recombinant factors in patients with hemophilia A [7].

We believe that DAA treatment reduces the need for rFVIIa by

lowering inhibitor levels through both suppression of hepatitis

C and modulation of cytokines.

Figure 1. Graph demonstrating inhibitor levels and factor VIIa dosage during direct-acting antiviral drug treatment.

146


Turk J Hematol 2022;39:144-151

LETTERS TO THE EDITOR

To the best of our knowledge, this case study is the first to report

reduced inhibitor levels and a decreased need for rFVIIa after

DAA treatment. This treatment may reduce inhibitor titers. The

treatment of hepatitis C may also play a role. We hope that our

findings will facilitate further studies involving larger numbers

of patients.

Keywords: FVIII deficiency, Hepatitis C, Inhibitor

Anahtar Sözcükler: FVIII eksikliği, Hepatit C, İnhibitör

Ethics

Informed Consent: Received.

Authorship Contributions

Surgical and Medical Practices: M.H.A., E.T., E.K.; Concept:

M.H.A.; Design: M.H.A.; Data Collection or Processing: M.H.A.;

Analysis or Interpretation: M.H.A., E.T., E.K.; Literature Search:

M.H.A., E.T., E.K.; Writing: M.H.A., E.T., E.K.

Conflict of Interest: The authors of this paper have no conflicts

of interest, including specific financial interests, relationships,

and/or affiliations relevant to the subject matter or materials

included.

References

1. Ljung RCR. How I manage patients with inherited haemophilia A and B and

factor inhibitors. Br J Haematol 2018;180:501-510.

2. Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a

systematic review. Haemophilia 2003;9:418-435.

3. Srivastava A, Santagostino E, Dougall A, Kitchen S, Sutherland M, Pipe

SW, Carcao M, Mahlangu J, Ragni MV, Windyga J, Llinás A, Goddard NJ,

Mohan R, Poonnoose PM, Feldman BM, Lewis SZ, van den Berg HM, Pierce

GF; WFH Guidelines for the Management of Hemophilia Panelists and Co-

Authors. WFH guidelines for the management of hemophilia, 3rd edition.

Haemophilia 2020;26:1-158.

4. Martins ML, Chaves DG, da Silva-Malta MCF, Bolina-Santos E, Barbosa-

Stancioli EF, Carmo RA. Hepatitis C and history of FVIII inhibitor

development in a long-term cohort of Brazilian patients with haemophilia

A. Haemophilia 2020;26:130-133.

5. Bolina-Santos E, Chaves DG, da Silva-Malta MCF, Carmo RA, Barbosa-

Stancioli EF, Lobato Martins M. HCV infection in hemophilia A patients

is associated with altered cytokines and chemokines profile and might

modulate the levels of FVIII inhibitor. J Med Virol 2022;94:683-691.

6. Holmes JA, Thompson AJ. Interferon-free combination therapies for the

treatment of hepatitis C: current insights. Hepat Med 2015;7:51-70.

7. Singh G, Sass R, Alamiry R, Zein N, Alkhouri N. Hepatitis C treatment

with triple therapy in a patient with hemophilia A. World J Clin Cases

2013;16:106-107.

Financial Disclosure: The authors declared that this study

received no financial support.

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Memiş Hilmi Atay, M.D., Ondokuz Mayıs University Faculty of

Medicine, Department of Internal Medicine, Division of Hematology, Samsun, Turkey

Phone : +90 530 468 15 59

E-mail : draatay@gmail.com ORCID: orcid.org/0000-0001-9666-6955

Received/Geliş tarihi: March 2, 2022

Accepted/Kabul tarihi: April 8, 2022

DOI: 10.4274/tjh.galenos.2022.2022.0085

147


LETTERS TO THE EDITOR

Turk J Hematol 2022;39:144-151

Spontaneous Remission in Paroxysmal Nocturnal Hemoglobinuria:

An Extremely Rare Case

Paroksismal Noktürnal Hemoglobinüride Spontan Remisyon: Çok Nadir Bir Olgu

Özgür Mehtap,

Ayfer Gedük

Kocaeli University Faculty of Medicine, Department of Hematology, Kocaeli, Turkey

To the Editor,

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired

clonal disease with the main clinical manifestations of

hemolytic anemia, bone marrow failure, and thrombophilia [1].

While it was previously reported that spontaneous remission

develops in 15%-30% of patients with PNH, this rate is as low

as 3% in recent studies [2,3,4,5]. This inconsistency between

results is probably due to the differences in the definitions of

“spontaneous remission” used in these studies and the analysis

methods applied. Herein, we present a rare case of aplastic

anemia/PNH overlap syndrome where the patient achieved

remission under immunosuppressive and anticomplement

therapy.

In 2012, a 24-year-old female patient was admitted to our

hospital due to severe fatigue. Pancytopenia was detected in

the complete blood count. Biochemical analysis was normal,

except for a slight increase in lactate dehydrogenase and a low

haptoglobin level. Bone marrow biopsy revealed decreased bone

marrow cellularity (40%) and relative erythroid hyperplasia. A

flow cytometric evaluation revealed a PNH granulocyte clone

size of 56.3%. The patient was started on eculizumab treatment,

which continued for 6 years. PNH clone percentages and clinical

parameters over the years are shown in Table 1. The PNH

clone size gradually decreased from 56.3% to 12.96% under

eculizumab treatment. Repeated bone marrow biopsy revealed

Table 1. Paroxysmal nocturnal hemoglobinuria clone percentages and clinical parameters over the years.

Type II erythrocytes, % Type III erythrocytes, % Total erythrocytes, % Monocytes, % Granulocytes, %

2012 2.75 11.63 14.38 48 56.3

2013 1.18 19.5 21.3 51.6 55

2014 1 13.9 14.9 40.3 46.5

2015 0.75 13 13.75 42.4 45

2016 0.28 12.56 12.84 48.5 42.6

2017 0.10 8.9 9 24.5 31.76

2018 10.95 10.53 21.48 11.05 12.96

2019 1.81 0.11 1.92 4.52 4.86

2020 1.68 0.04 1.72 4.26 3.58

2021 0.1 1.1 1.2 1.39 0.72

Neutrophils, /mm 3 Hemoglobin, g/dL Platelets, /mm 3 LDH, NR: 135-225 IU/L

2012 950 8.9 44,000 297 8

2013 1380 9.26 90,300 123 77

2014 1340 9.6 107,000 144 63

2015 4750 11.2 90,300 129 97

2016 1730 13.3 89,800 152 93

2017 1513 10.8 82,900 146 81

2018 695 10.67 48,700 180 138

2019 1634 11.13 95,000 150 99

2020 1896 11.89 102,000 187 118

2021 2119 13.69 123,700 171 106

LDH: Lactate dehydrogenase; NR: normal range.

Haptoglobin,

NR: 30-200 mg/dL

148


Turk J Hematol 2022;39:144-151

LETTERS TO THE EDITOR

a decrease in cellularity to 35% and, due to the tendency of

a decreasing platelet count, cyclosporine was added to the

treatment at 200 mg/day. After 1 year of combined treatment,

the pancytopenia was resolved, PNH clone size had decreased

to 4.86%, and follow-up bone marrow biopsy showed an

increase in cellularity (65%). With these results, eculizumab

and cyclosporine treatment was discontinued in 2019. The PNH

clone size of the patient, who is still asymptomatic 27 months

after the cessation of treatment, continues to shrink. During the

entire clinical course, the patient did not need transfusions and

no complications related to PNH developed.

The disappearance of the PNH clone may be due to various

causes, such as complete recovery, deepening of bone

marrow aplasia, or transformation to leukemia. However, the

underlying mechanisms and the reasons for complete recovery

are unknown. One of the hypotheses put forward is that

clones of cells affected by PNH have a limited lifespan like

normal somatic cells [3]. There are also cases in the literature

that draw attention to the relationship between pathological

PNH clones and bone marrow environmental conditions [6,7].

Recent studies have shown that PNH is a multiclonal disease

and hosts additional somatic mutations that result in a complex

hierarchical clonal architecture similar to that observed in

myeloid neoplasms. Thus, it has been suggested that remission

of PNH may occur through the emergence of a new dominant

clone carrying multiple somatic mutations rather than

restoration of normal hematopoiesis [5,8,9]. As a result, the

highly variable clinical spectrum of PNH is also reflected in cases

of remission. Understanding the underlying pathophysiology

of this extremely rare condition will form the basis for the

development of curative treatments for the disease.

Keywords: Aplastic anemia, Paroxysmal nocturnal

hemoglobinuria, Eculizumab, Cyclosporine

Anahtar kelimeler: Aplastik anemi, Paroksismal noktürnal

hemoglobinüri, Ekulizumab, Siklosporin

Analysis or Interpretation: Ö.M, A.G.; Literature Search: Ö.M,

A.G.; Writing: Ö.M, A.G.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Parker C, Omine M, Richards S, Nishimura J, Bessler M, Ware R, Hillmen

P, Luzzatto L, Young N, Kinoshita T, Rosse W, Socié G; International PNH

Interest Group. Diagnosis and management of paroxysmal nocturnal

hemoglobinuria. Blood 2005;106:3699-3709.

2. Muñoz-Linares C, Ojeda E, Forés R, Pastrana M, Cabero M, Morillo D, Bautista

G, Baños I, Monteserín C, Bravo P, Jaro E, Cedena T, Steegmann JL, Villegas A,

Cabrera JR. Paroxysmal nocturnal hemoglobinuria: a single Spanish center’s

experience over the last 40 yr. Eur J Haematol 2014;93:309-319.

3. Hillmen P, Lewis SM, Bessler M, Luzzatto L, Dacie JV. Natural history of

paroxysmal nocturnal hemoglobinuria. N Engl J Med 1995;333:1253-1258.

4. Korkama ES, Armstrong AE, Jarva H, Meri S. Spontaneous remission in

paroxysmal nocturnal hemoglobinuria-return to health or transition into

malignancy? Front Immunol 2018;9:1749.

5. Gurnari C, Pagliuca S, Kewan T, Bahaj W, Mori M, Patel BJ, Visconte V,

Maciejewski JP. Is nature truly healing itself? Spontaneous remissions in

paroxysmal nocturnal hemoglobinuria. Blood Cancer J 2021;11:187.

6. Pulini S, Marando L, Natale A, Pascariello C, Catinella V, Del Vecchio L,

Risitano AM, Fioritoni G. Paroxysmal nocturnal hemoglobinuria after

autologous stem cell transplantation: extinction of the clone during

treatment with eculizumab - pathophysiological implications of a unique

clinical case. Acta Haematol 2011;126:103-109.

7. Hakim F, Childs R, Balow J, Cowan K, Zujewski J, Gress R. Development

of paroxysmal nocturnal hemoglobinuria after chemotherapy. Blood

1996;88:4725-4726.

8. Shen W, Clemente MJ, Hosono N, Yoshida K, Przychodzen B, Yoshizato

T, Shiraishi Y, Miyano S, Ogawa S, Maciejewski JP, Makishima H. Deep

sequencing reveals stepwise mutation acquisition in paroxysmal nocturnal

hemoglobinuria. J Clin Invest 2014;124:4529-4538.

9. Babushok DV, Stanley N, Xie HM, Huang H, Bagg A, Olson TS, Bessler

M. Clonal replacement underlies spontaneous remission in paroxysmal

nocturnal haemoglobinuria. Br J Haematol 2017;176:487-490.

Authorship Contributions

Surgical and Medical Practices: Ö.M, A.G.; Concept: Ö.M, A.G.;

Design: Ö.M, A.G.; Data Collection or Processing: Ö.M, A.G.;

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Ayfer Gedük, M.D., Kocaeli University Faculty of Medicine,

Department of Hematology, Kocaeli, Turkey

E-mail : ayfergeduk@hotmail.com ORCID: orcid.org/0000-0002-5603-1178

Received/Geliş tarihi: December 7, 2021

Accepted/Kabul tarihi: January 27, 2022

DOI: 10.4274/tjh.galenos.2022.2021.0672

149


LETTERS TO THE EDITOR

Turk J Hematol 2022;39:144-151

Hematopoietic Stem Cell Transplantation to a Patient with Acute

Myeloid Leukemia from a Sibling Donor Positive for SARS-CoV-2

by RT-PCR Test

SARS-CoV-2 RT-PCR Testi Pozitif Kardeş Donörden Akut Myeloid Lösemili Bir Hastaya

Hematopoetik Kök Hücre Nakli

Ahmet Koç 1,2 , Ömer Doğru 1,2 , Nurşah Eker 1,2 , Burcu Tufan Taş 1,2 , Rabia Emel Şenay 1,2

1Marmara University Faculty of Medicine, Department of Pediatric Hematology and Oncology, İstanbul, Turkey

2Marmara University Training and Research Hospital, Pediatric Bone Marrow Transplant Unit, İstanbul, Turkey

To the Editor,

Hematopoietic stem cell transplantation (HSCT) is the preferred

treatment modality in cases of high-risk pediatric acute myeloid

leukemia (AML). However, due to the ongoing coronavirus

disease-2019 (COVID-19) pandemic, there is a risk that stem cell

donors may be positive for SARS-CoV-2 by reverse transcription

polymerase chain reaction (RT-PCR) and that the ability to find

suitable donors will be impacted [1,2]. In addition, it has been

reported that the virus can be found in the blood of patients

infected with SARS-CoV-2 [3]. Stem cell collection is not

recommended by the European Society for Blood and Marrow

Transplantation or other international organizations when the

donor’s RT-PCR test is positive for SARS-CoV-2 [1,2]. However,

in the literature, cases of HSCT performed with SARS-CoV-2-

positive donors due to necessity have been reported for a small

number of patients [4,5,6]. In this letter, we present the case of

a patient with high-risk AML who underwent HSCT from a fully

HLA-matched 22-year-old sibling with SARS-CoV-2 positivity

by RT-PCR test. Considering that the COVID-19 pandemic is

still ongoing with the emergence of new mutations, we wanted

to share this experience with colleagues who may encounter

similar situations.

A 15-year-old girl with a diagnosis of AML-M4 was included

in the high-risk AML group due to monosomy-7 positivity. She

received the first four blocks of the AML-BFM 2019 treatment

protocol (cytarabine, idarubicin, and etoposide in the first

treatment block; high-dose cytarabine and mitoxantrone

in the second treatment block; cytarabine and idarubicin

in the third treatment block; and high-dose cytarabine and

mitoxantrone in the fourth treatment block) and good response

was obtained. Before transplantation, her bone marrow was in

complete remission morphologically. Minimal residual disease

as measured by flowcytometry was below 1%. Monosomy-7

disappeared after the first block of induction therapy and

was also negative before transplantation. A nasopharyngeal

swab from the donor before conditioning began was

negative for SARS-CoV-2 by RT-PCR. Intravenous busulfan

(3.2 mg/kg/day, days -7 to -4), fludarabine (30 mg/m 2 /day,

days -7 to -3), and melphalan (140 mg /m 2 /day, day -1) were

administered as myeloablative conditioning. However, the

donor’s RT-PCR on the day before the transplant was positive for

SARS-CoV-2. The donor was asymptomatic and did not receive

any medication for COVID-19. There was no chance of finding

another donor for the patient at this stage. The transplant was

delayed 1 day and the donor’s SARS-CoV-2 test performed 2 days

after the previous test was again found to be positive. Since the

patient had serious infectious conditions during AML treatment,

it was decided to proceed with the transplant considering that

the risk of serious infection would increase significantly if the

transplantation was not performed at this time and the bone

marrow remained aplastic.

Following all personal protection rules issued by all relevant

health institutions, CD34+ peripheral blood stem cells were

collected from the donor by apheresis and were transfused

to the patient on the same day without any manipulation.

The patient and her accompanying mother wore appropriate

personal protective equipment during the transplantation

and did not develop any fever or other COVID-related

symptoms. Cyclosporine A (3 mg/kg/day, intravenous) and

methotrexate (10 mg/kg/day, on days 1, 3, and 6) were used for

graft-versus-host disease prophylaxis. Ciprofloxacin,

voriconazole, and acyclovir were used for infection prophylaxis.

Neutrophil engraftment occurred on day 14 and platelet

engraftment on day 16. The patient underwent a weekly SARS-

CoV-2 test after transplantation and all results were negative.

Bone marrow examination showed 100% donor chimerism at

the end of the fourth week. The patient is currently alive and

healthy in the fifth month after transplantation.

150


Turk J Hematol 2022;39:144-151

LETTERS TO THE EDITOR

In conclusion, this case supports the reports in the literature

that hematopoietic stem cells from asymptomatic SARS-CoV-2-

positive donors may be safe.

Keywords: Acute myeloid leukemia, COVID-19, Hematopoietic

stem cell transplantation

Anahtar Sözcükler: Akut myeloid lösemi, COVID-19,

Hematopoietik kök hücre nakli

Informed Consent: Obtained.

Authorship Contributions

Surgical and Medical Practices: A.K., Ö.D., N.E., B.T.T., R.E.Ş.;

Concept: A.K., Ö.D., N.E., B.T.T., R.E.Ş.; Design: A.K., Ö.D., N.E.,

B.T.T., R.E.Ş.; Data Collection or Processing: A.K., Ö.D., N.E., B.T.T.,

R.E.Ş.; Analysis or Interpretation: A.K., Ö.D., N.E., B.T.T., R.E.Ş.;

Literature Search: A.K.; Writing: A.K., Ö.D., N.E., B.T.T., R.E.Ş.

Conflict of Interest: No conflict of interest was declared by the

authors.

Financial Disclosure: The authors declared that this study

received no financial support.

References

1. Ljungman P, Mikulska M, de la Camara R, Basak GV, Chabannon C, Corbacioglu

S, Duarte R, Dolstra H, Lankester AC, Mohty M, Montoto S, Murray J, de

Latour RG, Snowden JA, Yakoub-Agha I, Verhoeven B, Kröger N, Styczynski

J, for the European Society for Blood and Marrow Transplantation. The

challenge of COVID-19 and hematopoietic cell transplantation; EBMT

recommendations for management of hematopoietic cell transplant

recipients, their donors, and patients undergoing CAR T-cell therapy. Bone

Marrow Transplantation 2020;55:2071-2076.

2. Waghmare A, Abidi MZ, Boeckh M, Chemaly RF, Dadwal S, Boghdadly Z,

Kamboj M, Papanicolaou GA, Pergam SA, Shahid Z. Guidelines for COVID-19

management in hematopoietic cell transplantation and cellular therapy

recipients. Biol Blood Marrow Transplant 2020;26:1983-1994.

3. Chang L, Yan Y, Wang L. Coronavirus disease 2019: coronaviruses and blood

safety. Transfus Med Rev 2020;34:75-80.

4. Anurathapan U, Apiwattanakul N, Pakakasama S, Pongphitcha P,

Thitithanyanont A, Pasomsub E, Hongeng S. Hematopoietic stem cell

transplantation from an infected SARS-CoV2 donor sibling. Bone Marrow

Transplant 2020;55:2359-2360.

5. Lázaro Del Campo P, de Paz Arias R, Ramírez López A, de la Cruz Benito B,

Humala Barbier K, Sánchez Vadillo I, López de la Guía A, de Soto Álvarez T,

Jiménez Yuste V, Canales Albendea M. No transmission of SARS-CoV-2 in

a patient undergoing allogeneic hematopoietic cell transplantation from

a matched-related donor with unknown COVID-19. Transfus Apher Sci

2020;59:102921.

6. Leclerc M, Fourati S, Menouche D, Challine D, Maury S. Allogeneic

haematopoietic stem cell transplantation from SARS-CoV-2 positive

donors. Lancet Haematol 2021;8:e167-169.

©Copyright 2022 by Turkish Society of Hematology

Turkish Journal of Hematology, Published by Galenos Publishing House

Address for Correspondence/Yazışma Adresi: Ahmet Koç, Ph.D. Marmara University Faculty of Medicine,

Department of Pediatric Hematology and Oncology, İstanbul, Turkey

E-mail : ahmetkoc1@hotmail.com ORCID: orcid.org/0000-0001-7940-2640

Received/Geliş tarihi: March 2, 2022

Accepted/Kabul tarihi: April 26, 2022

DOI: 10.4274/tjh.galenos.2022.2022.0086

151


Advisory Board of This Issue (June 2022)

Ahmet Emre Eşkazan, Turkey

Ahmet Muzaffer Demir, Turkey

Akif Selim Yavuz, Turkey

Ana Planinc-Peraica, Croatia

Atilla Özkan, Turkey

Burhan Ferhanoğlu, Turkey

Elif Gülsüm Ümit, Turkey

Emin Kaya, Turkey

Emine Zengin, Turkey

Emre Tekgündüz, Turkey

Esra Yıldızhan, Turkey

Fergün Yılmaz, Turkey

Hale Ören, Turkey

Hasan Hashem, Jordan

İnci Alacacıoğlu, Turkey

İrfan Yavaşoğlu, Turkey

Lenka Besse, Switzerland

Massimo Martino, Italy

Mehmet Can Uğur, Turkey

Müge Sayitoğlu, Turkey

Mustafa Baydar, Turkey

Nadim Mahmud, USA

Neslihan Andıç, Turkey

Nükhet Tüzüner, Turkey

Ömür Gökmen Sevindik, Turkey

Ömür Kayıkçı, Turkey

Rejin Kebudi, Turkey

Rouslan Kotchetkov, Canada

Sinan Demircioğlu, Turkey

Stephanie Poulain, France

Tekin Aksu, Turkey

Theoni Kanellopoulou, Greece

Tuba Karapınar, Turkey

Tuğrul Elverdi, Turkey

Tülin Tiraje Celkan, Turkey

Ümit Yavuz Malkan, Turkey

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

Saved successfully!

Ooh no, something went wrong!