Quality of life in adolescent and young adult Hodgkin’s lymphoma survivors

Ye.G. Arshanskaya1,2, S.V. Semochkin2,3, and A.G. Rumyantsev2,3

1 S.P. Botkin City Clinical Hospital, Moscow, Russian Federation

2 Federal Clinical-and-Research Center of Pediatric Hematology, Oncology, and Immunology n.a. Dmitriy Rogachev, Moscow, Russian Federation

3 N.I. Pirogov Russian National Research Medical University, RF Ministry of Health, Moscow, Russian Federation


ABSTRACT

Background. Deterioration of quality of life (QoL) and late complications of antitumor therapy for Hodgkin’s lymphoma (HL) are the important medical issues, since they mostly relate to young patients with a high life-expectancy.

Objective. The study was to compare QoL of HL survivors versus healthy young adults.

Methods. 56 (22 males and 34 females) HL survivors with a median age of 27.5 (range 22–41) were evaluated. For the purpose of comparison, 94 (44 males and 50 females) healthy subjects with a median age of 28.0 (range 22–46) were enrolled into the study of QoL. All HL survivors were treated in our hospital according to the modified pediatric protocol DAL-HD-90 in 1997–2007. QoL was assessed using the Short Form 36 (SF-36) which enabled generating 8 separate scales and 2 final scores (0 = worst possible health, 100 = best possible health). All survivors were in complete remission of HL for ³ 5 years.

Results. The HL survivors had the lower scores than the normal controls according to all scales and SF-36. Statistically significant differences were found in: general health — 53.4 (95 %CI 47.8–59.1) vs. 72.3 (68.8–75.8; < 0,001), vitality — 54.7 (50.4–59.1) vs. 72.2 (69.3–75.2; < 0.001), and mental health — 57.4 (53.5–61.4) vs. 71.7 (68.6–74.8; < 0.001). The patients at the age ³ 18.5 years (ROC-curves; = 0.047) at the time of HL diagnosis had poorer QoL when compared to younger patients with respect to: general health — 48.3 (41.3–55.2) vs. 60.9 (51.6–70.2; = 0.027): vitality — 50.3 (44.7–55.9) vs. 61.1 (51.6–70.2; = 0.013). The patients with the unfavorable events including relapse (n = 6) and second malignancy (n = 2) showed the lowest scores of QoL, especially in physical role performance [34.4 (2.6–71.3) vs. 79.7 (77.8–89.6; = 0.002)] and emotional role performance [25.0 (7.5–57.5) vs. 77.8 (67.1–88.4; < 0.001)]. Duration of remission, age at the QoL evaluation, gender, therapy intensity (2, 4, or 6 cycles of primary chemotherapy plus radiotherapy), Ann-Arbor stages, bulky disease, current married status, and education levels showed no significant influence on the QoL parameters.

Conclusion. Long-term HL survivors had poorer physical and mental QoL than the general population of young adults. The age at the time of LH diagnosis ³ 18.5 years was associated with significantly reduced QoL. The relapsed HL and second malignancies were mostly associated with the deterioration of physical and emotional role functioning that may indicate uncertainty of patients about future well-being.


Keywords: Hodgkin’s lymphoma, quality of life, adolescents, young adults, DAL-HD-90, SF-36

Read in PDF (RUS)pdficon


REFERENCES

  1. Злокачественные новообразования в России в 2012 г. (заболева- емость и смертность). Под ред. А.Д. Каприна, В.В. Старинского, Г.В. Пе- тровой. М.: ФГБУ «МНИОИ им. П.А. Герцена» МЗ РФ, 2014. [Sostoyaniye onkologicheskoy pomoshchi naseleniyu Rossii v 2012 g. Pod red. A.D. Kaprina, V.V. Starinskogo, G.V. Petrovoy (Status of oncological care for population of Russia in 2012. Ed. by A.D.Kaprin, V.V. Starinsky, and G.V. Petrova). M.: FGBU MNIO im. P.A. Gertsena Minzdravsotsrazvitiya Rossii 2014.]
  2. Morton L.M., Wang S.S., Devesa S.S. et al. Lymphoma incidence patterns by WHO subtype in the United States, 1992–2001. Blood 2006; 107(1): 265–76.
  3. Румянцев А.Г., Птушкин В.В., Семочкин С.В. Пути улучшения ре- зультатов лечения злокачественных опухолей у подростков и молодых взрослых. Онкогематология 2011; 1: 20–30. [Rumyantsev A.G., Ptushkin V.V., Semochkin S.V. Approaches to improvement of treatment outcomes in adolescents and young adults with malignancies. Onkogematologiya 2011; 1: 20–30. (In Russ.)].
  4. Evens A.M., Hutchings M., Diehl V. Treatment of Hodgkin lymphoma: the past, present, and future. Nat. Clin. Pract. Oncol. 2008; 5(9): 543–56.
  5. Sasse S., Klimm B., Gorgen H. et al. Comparing long-term toxicity and efficacy of combined modality treatment including extended- or involved-field radiotherapy in early-stage Hodgkin’s lymphoma. Ann. Oncol. 2012; 23(11): 2953–9.
  6. Robison L.L., Green D.M., Hudson M. et al. Long-term outcomes of adult survivors of childhood cancer. Cancer 2005; 104(11): 2557–64.
  7. Shah A.B., Hudson M.M., Poquette C.A. et al. Long-term follow-up of patients treated with primary radiotherapy for supradiaphragmatic Hodgkin’s disease at St. Jude Children’s Research Hospital. Int. J. Radiat. Oncol. Biol. Phys. 1999; 44(4): 867–77.
  8. Феоктистов Р.И., Румянцева Ю.В., Абугова Ю.Г. и др. Результаты лечения детей и подростков с лимфомой Ходжкина: данные многоцентро- вого исследования. Онкогематология 2010; 1: 31–6. [Feoktistov R.I., Rumyantseva Yu.V., Abugova Yu.G., et al. Treatment outcomes in children and adolescents with Hodgkin’s lymphoma: data of multicenter study. Onkogematologiya 2010; 1: 31–6. (In Russ.)].
  9. Schellong G., Riepenhausen M., Bruch C. et al. Late valvular and other cardiac diseases after different doses of mediastinal radiotherapy for Hodgkin disease in children and adolescents: report from the longitudinal GPOH followup project of the German-Austrian DAL-HD studies. Pediatr. Blood Cancer 2010; 55(6): 1145–52.
  10. Демина Е.А., Пылова И.В., Шмаков Р.Г., Перилова Е.Е. Поздние ослож- нения терапии больных лимфомой Ходжкина. Совр. онкол. 2006; 1: 36–43. [Demina Ye.A., Pylova I.V., Shmakov R.G., Perilova Ye.Ye. Late complications of therapy in patients with Hodgkin’s lymphoma. Sovr. onkol. 2006; 1: 36–43. (In Russ.)]. 11. Ng A.K., LaCasce A., Travis L.B. Long-term complications of lymphoma and its treatment. J. Clin. Oncol. 2011; 29(14): 1885–92.
  11. Gil-Fernandez J., Ramos C., Tamayo T. et al. Quality of life and psychological well-being in Spanish long-term survivors of Hodgkin’s disease: results of a controlled pilot study. Ann. Hematol. 2003; 82(1): 14–8.
  12. Khimani N., Chen Y.H., Mauch P.M. et al. Influence of new late effects on quality of life over time in Hodgkin lymphoma Survivors: a longitudinal survey study. Ann. Oncol. 2013; 24(1): 226–30.
  13. Hjermstad M.J., Fossa S.D., Oldervoll L. et al. Fatigue in long-term Hodgkin’s disease survivors: a follow-up study. J. Clin. Oncol. 2005; 23(27): 6587–95.
  14. Ruffer J.U., Flechtner H., Tralls P. et al. Fatigue in long-term survivors of Hodgkin’s lymphoma; a report from the German Hodgkin Lymphoma Study Group (GHSG). Eur. J. Cancer 2003; 39(15): 2179–86.
  15. Новик А.А., Ионова Т.И. Руководство по исследованию качества жизни в медицине. Под ред. Ю.Л. Шевченко, 2-е изд. М.: Олма Медиагрупп, 2007. [Novik A.A., Ionova T.I. Rukovodstvo po issledovaniyu kachestva zhizni v meditsine. Pod red. Yu.L. Shevchenko, 2-e izd. (Manual on evaluation of quality of life in medicine. Ed by.: Yu.L. Shevchenko, 2nd ed.) M.: Olma Mediagrup, 2007]
  16. Давыдкин И.Л., Булгакова С.В., Шафиева И.А. Подходы к реаби- литации пациентов в Самарском областном межведомственном центре профилактики остеопороза. Аллергол. и иммунол. 2007; 8(1): 276. [Davydkin I.L., Bulgakova S.V., Shafiyeva I.A. Approaches to rehabilitation of patients in Samara regional interdisciplinary center for osteoporosis prevention. Allergol. i immunol. 2007; 8(1): 276. (In Russ.)].
  17. Semochkin S.V., Arshanskaya E.G., Bobkova M.M., Rumiantsev A.G. A long-term follow-up report on the modified pediatric protocol DAL-HD-90 for adolescents and young adults with Hodgkin lymphoma. Pediatr. Blood Cancer 2012; 59(6): 1042.
  18. Семочкин С.В., Лория С.С., Румянцев А.Г., Сотников В.М. Лечение лимфомы Ходжкина у подростков и молодых взрослых. Онкогематология 2008; 1: 18–26. [Semochkin S.V., Loriya S.S., Rumyantsev A.G., Sotnikov V.M. Management of Hodgkin’s lymphoma in adolescents and young adults. Onkogematologiya 2008; 1: 18–26. (In Russ.)].
  19. Ware J.E., Kosinski M. Interpreting SF-36 summary health measures: a response. Qual. Life Res. 2001; 10(5): 405–13.
  20. Van Tulder M.W., Aaronson N.K., Bruning P.F. The quality of life of longterm survivors of Hodgkin’s disease. Ann. Oncol. 1994; 5(2): 153–8.
  21. Loge J.H., Abrahamsen A.F., Ekeberg O., Kaasa S. Reduced healthrelated quality of life among Hodgkin’s disease survivors: a comparative study with general population norms. Ann. Oncol. 1999; 10(1): 71–7.
  22. Mols F., Vingerhoets A.J., Coebergh J.W. et al. Better quality of life among 10–15 year survivors of Hodgkin’s lymphoma compared to 5–9 year survivors: a population-based study. Eur. J. Cancer 2006; 42(16): 2794–801.

Determination of B-cell clonality in Hodgkin’s lymphoma

Yu.V. Sidorova, N.V. Ryzhikova, S.Yu. Smirnova, E.E. Nikulina, B.V. Biderman, A.M. Kovrigina, T.N. Moiseeva, N.N. Sharkunov, and A.B. Sudarikov

Hematology Research Center, Moscow, Russian Federation


ABSTRACT

B-cell origin of Hodgkin’s lymphoma was demonstrated using microdissection and single cell PCR of Reed-Sternberg and Hodgkin cells (R. Kuppers et al., 1994). We assessed B-cell clonality in the biopsy samples of 35 patients with Hodgkin’s lymphoma without microdissection. B-cell clonality was evaluated using PCR amplification by IGH (FR1, FR2, FR3) and IGK (Vk-Jk, Vk/intron-Kde) gene rearrangements with multiplex BIOMED-2 primer sets and subsequent fragment analysis using ABI PRISM 3130 Genetic Analyzer (Applied Biosystems). Clonality was found in 11 out of 35 (31,5 %) formalin fixed paraffin-embedded (FFPE) lymph node specimens from patients with Hodgkin’s lymphoma. In 11 cases when both FFPE and fresh frozen samples were available, we observed the similar results with the specimens of both types. No correlation was found between the presence of B-cell clones and age, histological type of Hodgkin lymphoma, type of tumor cell growth (syncytial or diffuse), number of eosinophils in tissues, or CD20/CD15 expression on the surface of tumor cells. The high incidence of B-cell clonality determined in Hodgkin’s lymphoma biopsy samples makes the B-cell clonality assay unsuitable for differential diagnosis between Hodgkin’s lymphoma and B-cell lymphomas.


Keywords: Hodgkin’s lymphoma, B-cell clonality, PCR, immunoglobulin gene rearrangements.

Read in PDF (RUS)pdficon


REFERENCES

  1. Ковригина А.М., Пробатова Н.А. Лимфома Ходжкина и крупноклеточная лимфома. М.: МИА, 2007. [Kovrigina A.M., Probatova N.A. Limfoma Khodzhkina i krupnokletochnye limfomy (Hodgkin’s lymphoma and large-cell lymphomas). M.: MIA, 2007: 212.]
  2. Harris N.L., Jaffe E.S., Stein H. et al. A Revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood 1994; 84: 1361–92.
  3. Harris N.L., Jaffe E.S., Diebold J. et al. The World Health Organization Classification of Neoplastic Diseases of the Hematopoietic and Lymphoid Tissues. Ann. Oncol. 1999; 10: 1419–32.
  4. Kuppers R., Rajewsky K., Zhao M. et al. Hodgkin disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development. Proc. Natl. Acad. Sci. U S A 1994; 91: 10962–6.
  5. Marafioti T., Hummel M., Foss H.-D. et al. Hodgkin and Reed-Sternberg cells represent an expansion of a single clone originating from a germinal centre B-cell with functional immunoglobulin gene rearrangements but defective immunoglobulin transcription. Blood 2000; 95: 1443–50.
  6. Re D., Muschen M., Ahmadi T. et al. Oct-2 and Bob-1 deficiency in Hodgkin and Reed Sternberg cells. Cancer Res. 2001; 61: 2080–4.
  7. Jundt F., Kley K., Anagnostopoulos I. et al. Loss of PU.1 expression is associated with defective immunoglobulin transcription in Hodgkin and ReedSternberg cells of classical Hodgkin disease. Blood 2002; 99: 3060–2.
  8. Ushmorov A., Ritz O., Hummel M. et al. Epigenetic silencing of the immunoglobulin heavy-chain gene in classical Hodgkin lymphoma-derived cell lines contributes to the loss of immunoglobulin expression. Blood 2004; 104: 3326–34.
  9. Theil J., Laumen H., Marafioti T. et al. Defective octamer-dependent transcription is responsible for silenced immunoglobulin transcription in ReedSternberg cells. Blood 2001; 97: 3191–6.
  10. Kanzler H., Kuppers R., Hansmann M.L. et al. Hodgkin and ReedSternberg cells in Hodgkin’s disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells. J. Exp. Med. 1996; 184: 1495–505.
  11. Brauninger A., Schmitz R., Bechtel D. et al. Molecular biology of Hodgkin’s and Reed/Sternberg cells in Hodgkin’s lymphoma. Int. J. Cancer 2006; 118: 1853–61.
  12. Angel C.A., Pringle J.H., Naylor J. et al. Analysis of antigen receptor genes in Hodgkin’s disease. J. Clin. Pathol. 1993; 46(4): 337–40.
  13. Kamel O.W., Chang P.P., Hsu F.J. et al. Clonal VDJ recombination of the immunoglobulin heavy chain gene by PCR in classical Hodgkin’s disease. Am. J. Clin. Pathol. 1995; 104: 419–23.
  14. Manzanal A., Santon A., Oliva H. et al. Evaluation of clonal immunoglobulin heavy chain rearrangements in Hodgkin’s disease using the polymerase chain reaction (PCR). Histopathology 1995; 27: 21–5.
  15. Chute D.J., Cousar J.B., Mahadevan M.S. et al. Detection of immunoglobulin heavy chain gene rearrangements in classic Hodgkin lymphoma using commercially available BIOMED-2 primers. Diagn. Mol. Pathol. 2008; 17(2): 65–72.
  16. Hebeda K.M., Van Altena M.C., Rombout P. et al. PCR clonality detection in Hodgkin lymphoma. J. Hemat. 2009; 2(1): 34–41.
  17. Burack W.R., Laughlin T.S., Friedberg J.W. et al. PCR assays detect B-lymphocyte clonality in formalin-fixed, paraffin-embedded specimens of classical Hodgkin lymphoma without microdissection. Am. J. Clin. Pathol. 2010; 134(1): 104–11.
  18. Wu L., Patten N., Yamashiro C.T., Chui B. Extraction and amplification of DNA from formalin-fixed, paraffin-embedded tissues. Appl. Immunohistochem. Mol. Morphol. 2002; 10: 269–74.
  19. Coombs N.J., Gough A.C., Primrose J.N. Optimisation of DNA and RNA extraction from archival formalin-fixed tissue. Nucl. Acids Res. 1999; 27: e12.
  20. Sidorova J.V., Biderman B.V., Nikulina E.E., Sudarikov A.B. A simple and efficient method for DNA extraction from skin and paraffin-embedded tissues applicable to T-cell clonality assays. Exp. Dermatol. 2012; 21(1): 57–60.
  21. Dongen J.J., Langerak A.W., Bruggemann M. et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia 2003; 17(12): 2257–317.
  22. Jarrett R.F. Viruses and lymphoma/leukaemia. J. Pathol. 2006; 208(2): 176–86.
  23. Al Saati T., Galoin S., Gravel S. et al. IgH and TcR-gamma gene rearrangements identified in Hodgkin disease by PCR demonstrate lack of correlation between genotype, phenotype, and Epstein–Barr virus status. J. Pathol. 1997; 181(4): 387–93.
  24. Manzanal A.I., Santon A., Acevedo A. et al. Molecular analysis of the IgH gene in 212 cases of Hodgkin’s disease: correlation of IgH clonality with the histologic and the immunocytochemical features. Mod. Pathol. 1997; 10(7): 679–85.

International prognostic score in advanced Hodgkin’s lymphoma

K.D. Kaplanov1, A.L. Shipaeva1, V.A. Vasil’yeva1, E.G. Gemdjian2, I.V. Matveeva1, L.S. Tregubova1, T.U. Klitochenko1, K.V. Demidenko1, O.B. Kalashnikova1, G.U. Vyskub1, O.E. Golubeva1, O.V. Levina1, V.A. Orlov1, and E.A. Demina3

1 Volgograd Regional Oncology Clinic #1, Volgograd, Russian Federation

2 Hematology Research Center, RF Ministry of Health, Moscow, Russian Federation

3 N.N. Blokhin Russian Cancer Research Center, RAMS, Moscow, Russian Federation


ABSTRACT

Since chemotherapy of Hodkgin’s lymphoma was introduced in early 60s, it has undergone fundamental changes that were associated with dramatic improvement in the disease prognosis. Currently, the various intensive modifications of original BEACOPP, such as BEACOPP-14 and escalate BEACOPP, are among the most widely used for treatment of advanced Hodkgin’s lymphoma.

Initially, the International Prognostic Score (IPS) was developed for patients treated with MOPP and MOPP-ABVD protocols. We suggest that due to the well-known changing value of the various prognostic signs with protocols of different intensity, the significance of IPS for BEACOPP-based therapy should be reconsidered.

One hundred seventy two patients with advanced Hodgkin’s lymphoma were included in our trial. All these patients were treated at the Hematology department of Volgograd Regional Oncology Clinic #1. Treatment options were as follows: 64 (37%), 84 (49%), and 24 (14%) patients received intensive BEACOPP-based, standard BEACOPP, or ABVD therapy, respectively. The final data presented are related to the period up to June 30, 2012.

We retrospectively evaluated the treatment outcomes for each IPS group. To distinguish the most significant prognostic signs from all six IPS factors, we studied the impact of each factor on treatment efficacy.

The greatest difference in overall 3- and 4-year survival was observed between the groups of patients with IPS 0–1 and ³ 2; for IPS 0–1, 3- and 4-year overall survival rate was 93%; for IPS ³ 2, 3- and 4-year overall survival rate was 81% and 75%, respectively (= 0.05). 3-year overall survival was significantly negatively affected by such factors as age over 45 (70% versus 87%, relative risk (RR) = 3.95% CI: 1.7–7, = 0.01) and albumin level < 40 g/L (79% versus 88%, RR= 2.8, 95% CI: 1.2–6.8, p = 0.02). Overall 3-year survival rate in males (= 91) and females (n = 81) was 80% and 88%, respectively (= 0.09). We found no effect on overall and freedom-from-treatment-failure survival (FFTF) of such factors as hemoglobin levels, lymphocyte count, leukocytes count, and IV stage disease. With respect to overall survival, multivariate analysis showed the greatest significance of age (relative risk, RR =3.6, 95% CI: 1.8–7, = 0.001) and albumin level (OR = 2.6, 95% CI: 1.1–6, = 0.036).


Keywords: Hodgkin’s lymphoma, international prognostic score (IPS), advanced stages, overall survival (OS), freedom-from-treatment-failure survival (FFTFS), BEACOPP, ABVD

Read in PDF (RUS)pdficon


REFERENCES

  1. Richardson S.E., McNamara C. The management of classical Hodgkin’s lymphoma: past, present, and future. Advant. Hematol. 2011; 2011: 1–17.
  2. Kuruvilla J. Standard therapy of advanced Hodgkin lymphoma. Hematol. Am. Soc. Hematol. Educ. 2009: 497–506.
  3. Federico M., Luminari S., Iannitto E. et al. ABVD compared with BEACOPP compared with CEC for initial treatment of patients with advanced Hodgkin’s lymphoma: Results from the HD2000 Gruppo Italiani perlo Studio dei Linfomi Trial. Clin. Oncol. 2009; 27: 805–11.
  4. Gianni A.M., Rambaldi A., Zinzani P. Comparable 3-year outcome following ABVD or BEACOPP first-line chemotherapy, plus pre-planned high-dose salvage, in advanced Hodgkin lymphoma: a randomized trial of the Michelagelo, GITIL and IIL cooperative groups. ASCO meeting Chicago, 2008. Abstract 8506.
  5. Byar D.P. Identification of prognostic factors. In: Cancer clinical trials. Methods and practice. Ed. by M.E. Buyse, M.J. Staquet, R.J. Sylvester. Oxford: Oxford University Press, 1988.
  6. Hasenclever D., Diehl V. A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s disease. N. Engl. J. Med. 1998; 339(21): 1506–14.
  7. R. Optimal Therapy of Advanced Hodgkin Lymphoma. ASH Education book. 2011: 310–316.
  8. Diehl V., Franklin J., Pfreundschuh M. et al. Standard and increased-dose BEACOPP chemotherapy compared with COPP-ABVD for advanced Hodgkin’s disease. Engl. J. Med. 2003; 348(24): 2386–95.
  9. Wagstaff J., Gregory W.M., Swindell R. et al. Prognostic factors for survival in stage IIIB and IV Hodgkin’s disease: a multivariate analyses comparing two specialist treatment centers. J. Cancer. 1988; 58: 487–92.
  10. Wagstaff J., Steward W., Jones M. et al. Factors affecting remission and survival in patients with advanced Hodgkin’s disease treated with MVPP. Oncol. 1986; 4: 135–47.
  11. Straus D.J., Gaynor J.J., Myers J. et al. Prognostic factors among 185 adults with newly diagnosed advanced Hodgkin’s disease treated alternating potentially noncross-resistant chemotherapy and intermediate-dose radiation therapy. Clin. Oncol. 1990; 8: 1173–86.
  12. Proctor S.J., Taylor P., Mackie M.J. et al. A numerical prognostic index for clinical use in identification of poor-risk patients with Hodgkin’s disease at diagnosis. The Scotland and Newcastle Lymphoma Group (SNLG) Therapy Working Party. Lymphoma 1992; 7 (Suppl.): 17–20.
  13. Low S.E., Horsman J.M., Walters S.J. et al. Risk-adjusted prognostic models for Hodgkin’s disease (HD) and grade II non- Hodgkin’s lymphoma (NHL II): validation on 6728 British National Lymphoma Investigation patients. J. Hematol. 2003; 120: 277–80.
  14. Gobbi P.G., Comelli M., Grignani G.E. et al. Estimate of expected survival at diagnosis in Hodgkin’s disease: a means of weighting prognostic factors and a tool for treatment choice and clinical research. A report from the International Database on Hodgkin’s Disease (IDHD). Hematologica 1994; 79: 241–55.
  15. Moccia A.A., Donaldson J., Chhanabhai M. et al. International Prognostic Score in Advanced- Stage Hodgkin’s Lymphoma: Altered Utility in the Modern Era. J. Clin. 2012; 30: 3383–8.
  16. Капланов К.Д., Шипаева А.Л., Васильева В.А. и др. Эффективность программ химиотерапии первой линии при различных стадиях лимфомы Ходжкина. Клин. онкогематол. 2012; 1: 22–9. [Kaplanov K.D., Shipayeva A.L., Vasilyeva V.A. i dr. Effektivnost programm khimioterapii pervoy linii pri razlichnykh stadiyakh limfomy Khodzhkina (Efficacy of first-line chemotherapy programs at various stages of Hodgkin’s disease). onkogematol. 2012; 1: 22–9.]

 

Hodgkin’s lymphoma and male fertility disorders

A.А. Vinokurov

Federal Clinical-and-Research Center of Pediatric Hematology, Oncology, and Immunology n.a. Dmitriy Rogachev, Moscow, Russian Federation


ABSTRACT

This literature review is focused on the mechanisms of the impact that cytotoxic drugs cause in male reproductive cells and the rate of infertility induced by the various regimens of combined chemotherapy for Hodgkin’s lymphoma. The specific endocrine abnormalities associated with antitumor treatment are described in detail and, also, routine and experimental methods for fertility preservation in cancer patients are presented.


Keywords: Hodgkin’s lymphoma, male infertility, sperm cryopreservation, male fertility preservation.

Read in PDF (RUS)pdficon


REFERNCES

  1. Steliarova-Foucher E., Stiller C., Kaatsch P. et al. Geographical patterns and time trends of cancer incidence and survival among children and adolescents in Europe since the 1970s (the ACCIS project): an epidemiological study. Lancet 2004; 364(9451): 2097–105.
  2. Ferlay J., Shin H.R., Bray F. et al. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer 2010; 127(12): 2893–917.
  3. Parkin D.M., Bray F., Ferlay J., Pisani P. Global cancer statistics, 2002. CA Cancer J. Clin. 2005; 55(2): 74–108.
  4. Давыдов М.И., Аксель Е.М. Заболеваемость злокачественными ново- образованиями населения России странах СНГ в 2004 г. Вестн. РОНЦ им. Н.Н. Блохина РАМН 2006; 17(3, прил. 1): 45–77. [Davydov M.I., Aksel Ye.M. Zabolevayemost zlokachestvennymi novoobrazovaniyami naseleniya Rossii stranakh SNG v 2004 g (Cancer incidence in population of Russia and CIS in 2004. In: Bullet. of N.N.Blokhin RCC, RAMS). Vestn. RONTS im. N.N. Blokhina RAMN 2006; 17(3, pril. 1): 45–77.]
  5. Чиссов В.И., Старинский В.В., Петрова Г.В. Злокачественные ново- образования в России в 2010 г. (Заболеваемость и смертность). М.: ФГБУ «МНИОИ им. П.А. Герцена» МЗ РФ, 2012. [Chissov V.I., Starinskiy V.V., Petrova G.V. Zlokachestvennye novoobrazovaniya v Rossii v 2010 g. (Zabolevayemost i smertnost) (Cancer in Russia in 2010. (Incidence and mortality)). M.: FGBU «MNIOI im. P.A. Gertsena» MZ RF, 2012.]
  6. Evens A.M., Antillon M., Aschebrook-Kilfoy B., Chiu B.C. Racial disparities in Hodgkin’s lymphoma: a comprehensive population-based analysis. Ann. Oncol. 2012; 23(8): 2128–37.
  7. Miller B.A., Chu K.C., Hankey B.F., Ries L.A. Cancer incidence and mortality patterns among specific Asian and Pacific Islander populations in the U.S. Cancer Causes Control 2008; 19(3): 227–56.
  8. Волкова М.А. Клиническая онкогематология: Руководство для врачей, 2-е изд. М.: Медицина, 2007: 679–80. [Volkova M.A. Klinicheskaya onkogematologiya: Rukovodstvo dlya vrachey, 2-e izd (Clinical oncohematology: manual for medical practitioners. 2nd ed.). M.: Meditsina, 2007: 679–80.]
  9. Bleyer A., Viny A., Barr R. Cancer in 15- to 29-year-olds by primary site. Oncologist 2006; 11(6): 590–601.
  10. Armitage J.O. Early-stage Hodgkin’s lymphoma. N. Engl. J. Med. 2010; 363(7): 653–62.
  11. Hewitt M.W. Childhood Cancer survivorship: improving care and quality of life. Washington DC: The National Academy Press, 2003.
  12. Пивник А.В., Растригин Н.А., Моисеева Т.Н. и др. Результаты лечения лимфогранулематоза по протоколу МОРР-АВVD в сочетании с лучевой терапией (десятилетнее наблюдение). Тер. арх. 2006; 8: 57–62. [Pivnik A.V., Rastrigin N.A., Moiseyeva T.N. i dr. Rezultaty lecheniya limfogranulematoza po protokolu MORR-AVVD v sochetanii s luchevoy terapiyey (desyatiletneye nablyudeniye) (Outcomes of Hodgkin’s disease treated according to МОРР-АВVD protocol in combination with radiotherapy (10-year observations)). Ter. arkh. 2006; 8: 57–62.]
  13. Donaldson S.S., Link M.P. Combined modality treatment with low-dose radiation and MOPP chemotherapy for children with Hodgkin’s disease. J. Clin. Oncol. 1987; 5(5): 742–9.
  14. Куприна И.В. Состояние щитовидной железы и особенности липид- ного обмена у больных лимфогранулематозом молодого и среднего возраста после комбинированной химиолучевой терапии: Автореф. дис. … канд. мед. наук. М., 2008.
  15. Kuprina I.V. Sostoyaniye shchitovidnoy zhelezy i osobennosti lipidnogo obmena u bolnykh limfogranulematozom molodogo i srednego vozrasta posle kombinirovannoy khimioluchevoy terapii: Avtoref. dis. … kand. med. nauk (State¼: of thyroid gland and lipid metabolism in young and middle-aged patients with Hodgkin’s disease after combination chemoradiotherapy. Author’s summary of dissertation for the degree of PHD). M., 2008.
  16. Насибов О.М. Фиброз легких, кардиопатии и вторичные опухоли у канд.¼лиц в длительной ремиссии лимфогранулематоза: Автореф. дис. мед. наук. М., 2000. [Nasibov O.M. Fibroz legkikh, kardiopatii i vtorichnye opukholi u lits v dlitelnoy remissii limfogranulematoza: Avtoref. dis. … kand. med. nauk (Pulmonary fibrosis, cardiopathies and secondary tumors during Hodgkin’s disease in long remission. Author’s summary of dissertation for the degree of PHD). M., 2000.]
  17. Шилин Д.Е., Пивник А.В., Расстригин Н.А., Игнашина Е.В., Марголин О.В. Профилактика репродуктивных нарушений, возникающих в процессе химиотерапии у женщин детородного возраста, страдающих болезнью Ходжкина. Тер. арх. 1998; 7: 49–53. [Shilin D.Ye., Pivnik A.V., Rasstrigin N.A., Ignashina Ye.V., Margolin O.V. Profilaktika reproduktivnykh narusheniy, voznikayushchikh v protsesse khimioterapii u zhenshchin detorodnogo vozrasta, stradayushchikh boleznyu Khodzhkina (Prevention of chemotherapy-related reproductive disorders in women of reproductive age with Hodgkin’s disease). Ter. arkh. 1998; 7: 49–53.]
  18. Schrader M., Muller M., Straub B., Miller K. The impact of chemotherapy on male fertility: a survey of the biologic basis and clinical aspects. Reprod. Toxicol. 2001; 15(6): 611–7.
  19. Myrehaug S., Pintilie M., Yun L. et al. A population-based study of cardiac morbidity among Hodgkin lymphoma patients with preexisting heart disease. Blood 2010; 116(13): 2237–40.
  20. Meistrich M.L., Vassilopoulou-Sellin R., Lipshultz L.I. Adverse effects of treatment: gonadal dysfunction, 7th ed. Philadelphia: Lippincott Williams & Wilkins, 2005.
  21. Tilmann C., Capel B. Cellular and molecular pathways regulating mammalian sex determination. Rec. Prog. Horm. Res. 2002; 57: 1–18.
  22. Bendel-Stenzel M., Anderson R., Heasman J., Wylie C. The origin and migration of primordial germ cells in the mouse. Semin. Cell Dev. Biol. 1998; 9(4): 393–400.
  23. Meachem S., von Schonfeldt V., Schlatt S. Spermatogonia: stem cells with a great perspective. Reproduction 2001; 121(6): 825–34.
  24. Nieschlag E., Behre H. Andrology. Male Reproductive Health and Dysfunction, 3d ed. Berlin Heidelberg: Springer-Verlag, 2010: 35–6.
  25. Chuma S., Kanatsu-Shinohara M., Inoue K. et al. Spermatogenesis from epiblast and primordial germ cells following transplantation into postnatal mouse testis. Development 2005; 132(1): 117–22.
  26. Nayernia K., Drabent B., Meinhardt A. et al. Triple knockouts reveal gene interactions affecting fertility of male mice. Mol. Reprod. Dev. 2005; 70(4): 406–16.
  27. Creemers L.B., den Ouden K., van Pelt A.M., de Rooij D.G. Maintenance of adult mouse type A spermatogonia in vitro: influence of serum and growth factors and comparison with prepubertal spermatogonial cell culture. Reproduction 2002; 124(6): 791–9.
  28. Feng L., Chen Y., Dettin L. et al. Generation and in vitro differentiation of a spermatogonial cell line. Science 2002; 297(5580): 392–5.
  29. Geijsen N., Horoschak M., Kim K. et al. Derivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 2004; 427(6970): 148–54.
  30. Nagano M., Shinohara T., Avarbock M., Brinster R. Retrovirus-mediated gene delivery into male germ line stem cells. FEBS Lett. 2000; 475(1): 7–10.
  31. van Pelt A.M., Roepers-Gajadien H.L., Gademan I.S., Creemers L.B., de Rooij D.G. Establishment of cell lines with rat spermatogonial stem cell characteristics. Endocrinology 2002; 143(5): 1845–50.
  32. Clermont Y., Antar M. Duration of the cycle of the seminiferous epithelium and the spermatogonial renewal in the monkey Macaca arctoides. Am. J. Anat. 1973; 136(2): 153–65.
  33. Fouquet J.P., Dadoune J.P. Renewal of spermatogonia in the monkey (Macaca fascicularis). Biol. Reprod. 1986; 35(1): 199–207.
  34. Hermann B., Sukhwani M., Simorangkir D., Chu T., Orwig K. Molecular dissection of the male germ cell lineage identifies putative spermatogonial stem cells in rhesus macaques. Hum. Reprod. 2009; 24(7): 1704–16.
  35. Kluin P.M., Kramer M.F., de Rooij D.G. Testicular development in Macaca virus after birth. Int. J. Androl. 1983; 6(1): 25–43.
  36. Zhengwei Y., McLachlan R., Bremner W., Wreford N. Quantitative (stereological) study of the normal spermatogenesis in the adult monkey (Macaca fascicularis). J. Androl. 1997; 18(6): 681–7.
  37. de Rooij D.G. The spermatogonial stem cell niche. Microsc. Res. Tech. 2009; 72(8): 580–5.
  38. Dettin L., Ravindranath N., Hofmann M., Dym M. Morphological characterization of the spermatogonial subtypes in the neonatal mouse testis. Biol. Reprod. 2003; 69(5): 1565–71.
  39. Jahnukainen K., Ehmcke J., Soder O., Schlatt S. Clinical potential and putative risks of fertility preservation in children utilizing gonadal tissue or germline stem cells. Pediatr. Res. 2006; 59(4 Pt. 2): 40R–7R.
  40. Nieschlag E., Behre H. Andrology. Male Reproductive Health and Dysfunction, 3d ed. Berlin Heidelberg: Springer-Verlag, 2010: 16–7.
  41. Allenby G., Foster P.M., Sharpe R.M. Evidence that secretion of immunoactive inhibin by seminiferous tubules from the adult rat testis is regulated by specific germ cell types: correlation between in vivo and in vitro studies. Endocrinology 1991; 128(1): 467–76.
  42. Pineau C., Sharpe R.M., Saunders P.T., Gerard N., Jegou B. Regulation of Sertoli cell inhibin production and of inhibin alpha-subunit mRNA levels by specific germ cell types. Mol. Cell Endocrinol. 1990; 72(1): 13–22.
  43. Нишлаг Э., Бере Г. Андрология. Мужское здоровье и дисфункция репродуктивной системы, 2-е изд. М.: МИА, 2005: 40–1. [Nishlag E., Bere G. Andrologiya. Muzhskoye zdorovye i disfunktsiya reproduktivnoy sistemy, 2-e izd (Andrology. Men’s health and reproductive system dysfunctions. 2nd ed.). M.: MIA, 2005: 40–1.]
  44. Chung K., Irani J., Knee G. et al. Sperm cryopreservation for male patients with cancer: an epidemiological analysis at the University of Pennsylvania. Eur. J. Obstet. Gynecol. Reprod. Biol. 2004; 113(Suppl. 1): S7–11.
  45. Williams D., Karpman E., Sander J., Spiess P., Pisters L. Pretreatment semen parameters in men with cancer. J. Urol. 2009; 181(2): 736–40.
  46. Bahadur G., Ozturk O., Muneer A. et al. Semen quality before and after gonadotoxic treatment. Hum. Reprod. 2005; 20(3): 774–81.
  47. Gandini L., Lombardo F., Salacone P. et al. Testicular cancer and Hodgkin’s disease: evaluation of semen quality. Hum. Reprod. 2003; 18(4): 796–801.
  48. Rofeim O., Gilbert B. Normal semen parameters in cancer patients presenting for cryopreservation before gonadotoxic therapy. Fertil. Steril. 2004; 82(2): 505–6.
  49. Rueffer U., Breuer K., Josting A. et al. Male gonadal dysfunction in patients with Hodgkin’s disease prior to treatment. Ann. Oncol. 2001; 12(9): 1307–11.
  50. van der Kaaij M., Heutte N., van Echten-Arends J. et al. Sperm quality before treatment in patients with early stage Hodgkin’s lymphoma enrolled in EORTC-GELA Lymphoma Group trials. Haematologica 2009; 94(12): 1691–7.
  51. Marmor D., Elefant E., Dauchez C., Roux C. Semen analysis in Hodgkin’s disease before the onset of treatment. Cancer 1986; 57(10): 1986–7.
  52. Padron O.F., Sharma R.K., Thomas A.J. Jr., Agarwal A. Effects of cancer on spermatozoa quality after cryopreservation: a 12-year experience. Fertil. Steril. 1997; 67(2): 326–31.
  53. Viviani S., Ragni G., Santoro A. et al. Testicular dysfunction in Hodgkin’s disease before and after treatment. Eur. J. Cancer 1991; 27(11): 1389–92.
  54. Barr R.D., Clark D.A., Booth J.D. Dyspermia in men with localized Hodgkin’s disease. A potentially reversible, immune-mediated disorder. Med. Hypotheses 1993; 40(3): 165–8.
  55. Buch J.P., Kolon T.F., Maulik N., Kreutzer D.L., Das D.K. Cytokines stimulate lipid membrane peroxidation of human sperm. Fertil. Steril. 1994; 62(1): 186–8.
  56. Dousset B., Hussenet F., Daudin M., Bujan L., Foliguet B. Seminal cytokine concentrations (IL-1beta, IL-2, IL-6, sR IL-2, sR IL-6), semen parameters and blood hormonal status in male infertility. Hum. Reprod. 1997; 12(7): 1476–9.
  57. Fedder J., Ellerman-Eriksen S. Effect of cytokines on sperm motility and ionophore-stimulated acrosome reaction. Arch. Androl. 1995; 35(3): 173–85.
  58. Gruschwitz M.S., Brezinschek R., Brezinschek H.P. Cytokine levels in the seminal plasma of infertile males. J. Androl. 1996; 17(2): 158–63.
  59. Huleihel M., Lunenfeld E., Levy A., Potashnik G., Glezerman M. Distinct expression levels of cytokines and soluble cytokine receptors in seminal plasma of fertile and infertile men. Fertil. Steril. 1996; 66(1): 135–9.
  60. Schulte H.M., Bamberger C.M., Elsen H., Herrmann G., Bamberger A.M. Systemic interleukin-1 alpha and interleukin-2 secretion in response to acute stress and to corticotropin-releasing hormone in humans. Eur. J. Clin. Invest. 1994; 24(11): 773–7.
  61. Shimonovitz S., Barak V., Zacut D., Ever-Hadani P., Ben C.A. High concentration of soluble interleukin-2 receptors in ejaculate with low sperm motility. Hum. Reprod. 1994; 9(4): 653–5.
  62. Agarwal A., Allamaneni S.S. Disruption of spermatogenesis by the cancer disease process. J. Natl. Cancer Inst. Monogr. 2005; 34: 9–12.
  63. Ho G.T., Gardner H., De Wolf W.C, Loughlin K.R., Morgentaler A. Influence of testicular carcinoma on ipsilateral spermatogenesis. J. Urol. 1992; 148(3): 821–5.
  64. Skakkebaek N.E., Jorgensen N., Main K.M. et al. Is human fecundity declining? Int. J. Androl. 2006; 29(1): 2–11.
  65. Spitz S. The histological effects of nitrogen mustards on human tumors and tissues. Cancer 1948; 1(3): 383–98.
  66. Magelssen H., Brydoy M., Fossa S.D. The effects of cancer and cancer treatments on male reproductive function. Nat. Clin. Pract. Urol. 2006; 3(6): 312–22.
  67. Trottmann M., Becker A.J., Stadler T. et al. Semen quality in men with malignant diseases before and after therapy and the role of cryopreservation. Eur. Urol. 2007; 52(2): 355–67.
  68. Meistrich M.L., Finch M., da Cunha M.F., Hacker U., Au W.W. Damaging effects of fourteen chemotherapeutic drugs on mouse testis cells. Cancer Res. 1982; 42(1): 122–31.
  69. Dejaco C., Mittermaier C., Reinisch W. et al. Azathioprine treatment and male fertility in inflammatory bowel disease. Gastroenterology 2001; 121(5): 1048–53.
  70. Abu-Baker S.O. Gemcitabine impacts histological structure of mice testis and embryonic organs. Pak. J. Biol. Sci. 2009; 12(8): 607–15.
  71. Savkovic N., Green S., Pecevski J., Maric N. The effect of mitomycin on the fertility and the induction of meiotic chromosome rearrangements in mice and their first generation progeny. Can. J. Genet. Cytol. 1977; 19(3): 387–93.
  72. Colpi G.M., Contalbi G.F., Nerva F., Sagone P., Piediferro G. Testicular function following chemo-radiotherapy. Eur. J. Obstet. Gynecol. Reprod. Biol. 2004; 113(Suppl. 1): S2–6.
  73. Kirchhoff C. CD52 is the ‘major maturation-associated’ sperm membrane antigen. Mol. Hum. Reprod. 1996; 2(1): 9–17.
  74. Vogel E.W., Nivard M.J. International Commission for Protection Against Environmental Mutagens and Carcinogens. The subtlety of alkylating agents in reactions with biological macromolecules. Mutat. Res. 1994; 305(1): 13–32.
  75. Vogel E.W., Barbin A., Nivard M.J., Bartsch H. Nucleophilic selectivity of alkylating agents and their hypermutability in Drosophila as predictors of carcinogenic potency in rodents. Carcinogenesis 1990; 11(12): 2211–7.
  76. Angerer J., Ewers U., Wilhelm M. Human biomonitoring: state of the art. Int. J. Hyg. Environ. Health 2007; 210(3–4): 201–28.
  77. Ehrenberg L. Covalent binding of genotoxic agents to proteins and nucleic acids. IARC Sci. Publ. 1984; 59: 107–14.
  78. Волкова М.А. Клиническая онкогематология: Руководство для врачей, 2-е изд. М.: Медицина, 2007: 699–700.  [Volkova M.A. Klinicheskaya onkogematologiya: Rukovodstvo dlya vrachey, 2-e izd. (Clinical oncohematology: manual for medical practitioners. 2nd ed.). M.: Meditsina, 2007: 699–700.]
  79. Sanders J.E., Hawley J., Levy W. et al. Pregnancies following high-dose cyclophosphamide with or without high-dose busulfan or total-body irradiation and bone marrow transplantation. Blood 1996; 87(7): 3045–52.
  80. Marina S., Barcelo P. Permanent sterility after immunosuppressive therapy. Intern. J. Androl. 1979; 2: 6–13.
  81. Buchanan J.D., Fairley K.F., Barrie J.U. Return of spermatogenesis after stopping cyclophosphamide therapy. Lancet 1975; 2(7926): 156–7.
  82. da Cunha M.F., Meistrich M.L., Fuller L.M. et al. Recovery of spermatogenesis after treatment for Hodgkin’s disease: limiting dose of MOPP chemotherapy. J. Clin. Oncol. 1984; 2(6): 571–7.
  83. Jacob A., Barker H., Goodman A., Holmes J. Recovery of spermatogenesis following bone marrow transplantation. Bone Marrow Transplant. 1998; 22(3): 277–9.
  84. Hansen P.V., Trykker H., Helkjoer P.E., Andersen J. Testicular function in patients with testicular cancer treated with orchiectomy alone or orchiectomy plus cisplatin-based chemotherapy. J. Natl. Cancer Inst. 1989; 81(16): 1246–50.
  85. Ahmed S.R., Shalet S.M., Campbell R.H., Deakin D.P. Primary gonadal damage following treatment of brain tumors in childhood. J. Pediatr. 1983; 103(4): 562–5.
  86. Kulkarni S.S., Sastry P.S., Saikia T.K., Parikh P.M., Gopal R. Gonadal function following ABVD therapy for Hodgkin’s disease. Am. J. Clin. Oncol. 1997; 20(4): 354–7.
  87. Radford J.A., Clark S., Crowther D., Shalet S.M. Male fertility after VAPECB chemotherapy for Hodgkin’s disease and non-Hodgkin’s lymphoma. Br. J. Cancer 1994; 69: 379–81.
  88. Sieniawski M., Reineke T., Josting A. et al. Assessment of male fertility in patients with Hodgkin’s lymphoma treated in the German Hodgkin Study Group (GHSG) clinical trials. Ann. Oncol. 2008; 19(10): 1795–801.
  89. Hobbie W.L., Ginsberg J.P., Ogle S.K., Carlson C.A., Meadows A.T. Fertility in males treated for Hodgkins disease with COPP/ABV hybrid. Pediatr. Blood Cancer 2005; 44(2): 193–6.
  90. Marmor D., Duyck F. Male reproductive potential after MOPP therapy for Hodgkin’s disease: a long-term survey. Andrologia 1995; 27(2): 99–106.
  91. Whitehead E., Shalet S.M., Blackledge G., Todd I., Crowther D. The effects of Hodgkin’s disease and combination chemotherapy on gonadal function in the adult male. Cancer 1982; 49(3): 418–22.
  92. Viviani S., Santoro A., Ragni G., Bonfante V., Bonadonna G. Gonadal toxicity after combination chemotherapy for Hodgkin’s disease. Comparative results of MOPP vs ABVD. Eur. J. Cancer Clin. Oncol. 1985; 21(5): 601–5.
  93. Anselmo A.P., Cartoni C., Bellantuono P., Maurizi-Enrici R., Aboulkair N. Risk of infertility in patients with Hodgkin’s disease treated with ABVD vs MOPP vs ABVD/MOPP. Haematologica 1990; 75(2): 155–8.
  94. Shafford E.A., Kingston J.E., Malpas J.S. et al. Testicular function following the treatment of Hodgkin’s disease in childhood. Br. J. Cancer 1993; 68(6): 1199–204.
  95. Charak B.S., Gupta R., Mandrekar P. et al. Testicular dysfunction after cyclophosphamide-vincristine-procarbazine-prednisolone chemotherapy for advanced Hodgkin’s disease. A long-term follow-up study. Cancer 1990; 65(9): 1903–6.
  96. Puscheck E., Philip P.A., Jeyendran R.S. Male fertility preservation and cancer treatment. Cancer Treat. Rev. 2004; 30(2): 173–80.
  97. Chapman R.M., Sutcliffe S.B., Rees L.H., Edwards C.R., Malpas J.S. Cyclical combination chemotherapy and gonadal function. Retrospective study in males. Lancet 1979; 1(8111): 285–9.
  98. Brougham M.F., Kelnar C.J., Sharpe R.M., Wallace W.H. Male fertility following childhood cancer: current concepts and future therapies. Asian J. Androl. 2003; 5(4): 325–37.
  99. Thomson A.B., Critchley H.O., Kelnar C.J., Wallace W.H. Late reproductive sequelae following treatment of childhood cancer and options for fertility preservation. Best Pract. Res. Clin. Endocrinol. Metab. 2002; 16(2): 311–34.
  100. Viviani S., Bonfante V., Santoro A. et al. Long-term results of an intensive regimen: VEBEP plus involved-field radiotherapy in advanced Hodgkin’s disease. Cancer J. Sci. Am. 1999; 5(5): 275–82.
  101. Sieniawski M., Reineke T., Nogova L. et al. Fertility in male patients with advanced Hodgkin lymphoma treated with BEACOPP: a report of the German Hodgkin Study Group (GHSG). Blood 2008; 111(1): 71–6.
  102. Ferme C., Bastion Y., Lepage E. et al. The MINE regimen as intensive salvage chemotherapy for relapsed and refractory Hodgkin’s disease. Ann. Oncol. 1995; 6(6): 543–9.
  103. Bartlett N.L., Niedzwiecki D., Johnson J.L. et al. Gemcitabine, vinorelbine, and pegylated liposomal doxorubicin (GVD), a salvage regimen in relapsed Hodgkin’s lymphoma: CALGB 59804. Ann. Oncol. 2007; 18(6): 1071–9.
  104. Hansen P.V., Trykker H., Svennekjaer I.L., Hvolby J. Long-term recovery of spermatogenesis after radiotherapy in patients with testicular cancer. Radiother. Oncol. 1990; 18(2): 117–25.
  105. Meistrich M.L. Quantitative Correlation Between Testicular Stem Cell Survival, Sperm Production, and Fertility in the Mouse After Treatment With Different Cytotoxic Agents. J. Androl. 1982; 3(1): 58–68.
  106. van Alphen M.M., van de Kant H.J., de Rooij D.G. Repopulation of the seminiferous epithelium of the rhesus monkey after Х irradiation. Radiat. Res. 1988; 113(3): 487–500.
  107. Zhang Z., Shao S., Meistrich M.L. The radiation-induced block in spermatogonial differentiation is due to damage to the somatic environment, not the germ cells. J. Cell Physiol. 2007; 211(1): 149–58.
  108. Anserini P., Chiodi S., Spinelli S. et al. Semen analysis following allogeneic bone marrow transplantation. Additional data for evidence-based counselling. Bone Marrow Transplant. 2002; 30(7): 447–51.
  109. Hahn E.W., Feingold S.M., Simpson L., Batata M. Recovery from aspermia induced by low-dose radiation in seminoma patients. Cancer 1982; 50(2): 337–40.
  110. Bonadonna G., Santoro A., Viviani S., Lombardi C., Ragni G. Gonadal damage in Hodgkin’s disease from cancer chemotherapeutic regimens. Arch. Toxicol. Suppl. 1984; 7: 140–5.
  111. Muller H.L., Klinkhammer-Schalke M., Seelbach-Gobel B., Hartmann A.A., Kuhl J. Gonadal function of young adults after therapy of malignancies during childhood or adolescence. Eur. J. Pediatr. 1996; 155(9): 763–9.
  112. Tal R., Botchan A., Hauser R., Yogev L., Paz G. Follow-up of sperm concentration and motility in patients with lymphoma. Hum. Reprod. 2000; 15(9): 1985–8.
  113. van Dorp W., van Beek R.D., Laven J.S., Pieters R., de Muinck KeizerSchrama S.M. Long-term endocrine side effects of childhood Hodgkin’s lymphoma treatment: a review. Hum. Reprod. Update 2012; 18(1): 12–28.
  114. Heikens J., Behrendt H., Adriaanse R., Berghout A. Irreversible gonadal damage in male survivors of pediatric Hodgkin’s disease. Cancer 1996; 78(9): 2020–4.
  115. Aubier F., Flamant F., Brauner R., Caillaud J.M., Chaussain J.M. Male gonadal function after chemotherapy for solid tumors in childhood. J. Clin. Oncol. 1989; 7(3): 304–9.
  116. Ben Arush M.W., Solt I., Lightman A., Linn S., Kuten A. Male gonadal function in survivors of childhood Hodgkin and non-Hodgkin lymphoma. Pediatr. Hematol. Oncol. 2000; 17(3): 239–45.
  117. Dhabhar B.N., Malhotra H., Joseph R. et al. Gonadal function in prepubertal boys following treatment for Hodgkin’s disease. Am. J. Pediatr. Hematol. Oncol. 1993; 15(3): 306–10.
  118. Sherins R.J., Olweny C.L., Ziegler J.L. Gynecomastia and gonadal dysfunction in adolescent boys treated with combination chemotherapy for Hodgkin’s disease. N. Engl. J. Med. 1978; 299(1): 12–6.
  119. Green D.M., Brecher M.L., Lindsay A.N. et al. Gonadal function in pediatric patients following treatment for Hodgkin disease. Med. Pediatr. Oncol. 1981; 9(3): 235–44.
  120. Ash P. The influence of radiation on fertility in man. Br. J. Radiol. 1980; 53(628): 271–8.
  121. Lee S.J., Schover L.R., Partridge A.H. et al. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J. Clin. Oncol. 2006; 24(18): 2917–31.
  122. Kinsella T.J., Trivette G., Rowland J. et al. Long-term follow-up of testicular function following radiation therapy for early-stage Hodgkin’s disease. J. Clin. Oncol. 1989; 7(6): 718–24.
  123. Rowley M.J., Leach D.R., Warner G.A., Heller C.G. Effect of graded doses of ionizing radiation on the human testis. Radiat. Res. 1974; 59(3): 665–78.
  124. Howell S.J., Shalet S.M. Spermatogenesis after cancer treatment: damage and recovery. J. Natl. Cancer Inst. Monogr. 2005; 34: 12–7.
  125. Giwercman A., von der Maase H., Berthelsen J.G., Rorth M., Skakkebaek N.E. Localized irradiation of testes with carcinoma in situ: effects on Leydig cell function and eradication of malignant germ cells in 20 patients. J. Clin. Endocrinol. Metab. 1991; 73(3): 596–603.
  126. Hahn E.W., Feingold S.M., Nisce L. Aspermia and recovery of spermatogenesis in cancer patients following incidental gonadal irradiation during treatment: a progress report. Radiology 1976; 119(1): 223–5.
  127. Socie G., Salooja N., Cohen A. et al. Nonmalignant late effects after allogeneic stem cell transplantation. Blood 2003; 101(9): 3373–85.
  128. Howell S.J., Radford J.A., Ryder W.D., Shalet S.M. Testicular function after cytotoxic chemotherapy: evidence of Leydig cell insufficiency. J. Clin. Oncol. 1999; 17(5): 1493–8.
  129. Petersen P.M., Andersson A.M., Rorth M., Daugaard G., Skakkebaek N.E. Undetectable inhibin B serum levels in men after testicular irradiation. J. Clin. Endocrinol. Metab. 1999; 84(1): 213–5.
  130. Нишлаг Э., Бере Г.М. Андрология. Мужское здоровье и дисфункция репродуктивной системы, 2-е изд. М.: МИА, 2005: 32–4.
  131. [Nishlag E., Bere G.M. Andrologiya. Muzhskoye zdorovye i disfunktsiya reproduktivnoy sistemy, 2-e izd. (Andrology. Men’s health and reproductive system dysfunctions. 2nd ed.). M.: MIA, 2005: 32–4.]
  132. Bohring C., Krause W. Serum levels of inhibin B in men with different causes of spermatogenic failure. Andrologia 1999; 31(3): 137–41.
  133. van Beek R.D., Smit M., van den Heuvel-Eibrink M.M. et al. Inhibin B is superior to FSH as a serum marker for spermatogenesis in men treated for Hodgkin’s lymphoma with chemotherapy during childhood. Hum. Reprod. 2007; 22(12): 3215–22.
  134. Halder A., Fauzdar A., Kumar A. Serum inhibin B and follicle-stimulating hormone levels as markers in the evaluation of azoospermic men: a comparison. Andrologia 2005; 37(5): 173–9.
  135. Carreau S. Paracrine control of human Leydig cell and Sertoli cell functions. Folia Histochem. Cytobiol. 1996; 34(3–4): 111–9.
  136. Huhtaniemi I., Toppari J. Endocrine, paracrine and autocrine regulation of testicular steroidogenesis. Adv. Exp. Med. Biol. 1995; 377: 33–54.
  137. Castillo L.A., Craft A.W., Kernahan J., Evans R.G., Aynsley-Green A. Gonadal function after 12-Gy testicular irradiation in childhood acute lymphoblastic leukaemia. Med. Pediatr. Oncol. 1990; 18(3): 185–9.
  138. Hobbie W.L., Schwartz C.L. Endocrine late effects among survivors of cancer. Semin. Oncol. Nurs. 1989; 5(1): 14–21.
  139. Shalet S.M., Horner A., Ahmed S.R., Morris-Jones P.H. Leydig cell damage after testicular irradiation for lymphoblastic leukaemia. Med. Pediatr. Oncol. 1985; 13(2): 65–8.
  140. Orwig K.E., Schlatt S. Cryopreservation and transplantation of spermatogonia and testicular tissue for preservation of male fertility. J. Natl. Cancer Inst. Monogr. 2005; 34: 51–6.
  141. Res U., Res P., Kastelic D., Stanovnik M., Kmetec A. Birth after treatment of a male with seminoma and azoospermia with cryopreserved-thawed testicular tissue. Hum. Reprod. 2000; 15(4): 861–4.
  142. Dohle G.R., Colpi G.M., Hargreave T.B., Papp G.K., Jungwirth A. EAU guidelines on male infertility. Eur. Urol. 2005; 48(5): 703–11.
  143. Hirsch M., Lunenfeld B., Modan M., Ovadia J., Shemesh J. Spermarche the age of onset of sperm emission. J. Adolesc. Health Care 1985; 6(1): 35–9.
  144. Nielsen C.T., Skakkebaek N.E., Richardson D.W. et al. Onset of the release of spermatozoa (spermarche) in boys in relation to age, testicular growth, pubic hair, and height. J. Clin. Endocrinol. Metab. 1986; 62(3): 532–5.
  145. Kamischke A., Jurgens H., Hertle L., Berdel W.E., Nieschlag E. Cryopreservation of sperm from adolescents and adults with malignancies. J. Androl. 2004; 25(4): 586–92.
  146. Hagenas I., Jorgensen N., Rechnitzer C. et al. Clinical and biochemical correlates of successful semen collection for cryopreservation from 12–18-yearold patients: a single-center study of 86 adolescents. Hum. Reprod. 2010; 25(8): 2031–8.
  147. Bahadur G., Ling K.L., Hart R. et al. Semen quality and cryopreservation in adolescent cancer patients. Hum. Reprod. 2002; 17(12): 3157–61.
  148. Naysmith T.E., Blake D.A., Harvey V.J., Johnson N.P. Do men undergoing sterilizing cancer treatments have a fertile future? Hum. Reprod. 1998; 13(11): 3250–5.
  149. Park Y.S., Lee S.H., Song S.J., Jun J.H., Koong M.K. Influence of motility on the outcome of in vitro fertilization/intracytoplasmic sperm injection with fresh vs. frozen testicular sperm from men with obstructive azoospermia. Fertil. Steril. 2003; 80(3): 526–30.
  150. Cryopreservation of spermatozoa. Laboratory manual for the examination and processing of human semen, 5th ed. Geneva: World Health Organization press, 2010: 169–71.
  151. Perloff W.H., Steinberger E., Sherman J.K. Conception with human spermatozoa frosen by nirogen vapor technic. Fertil. Steril. 1964; 15: 501–4.
  152. David G., Czyglik F., Mayaux M.J., Schwartz D. The success of A.I.D. and semen characteristics: study on 1489 cycles and 192 ejaculates. Int. J. Androl. 1980; 3(6): 613–9.
  153. Clarke G.N., Bourne H., Hill P. et al. Artificial insemination and in-vitro fertilization using donor spermatozoa: a report on 15 years of experience. Hum. Reprod. 1997; 12(4): 722–6.
  154. Leibo S.P., Picton H.M., Gosden R.G. Cryopreservation of human spermatozoa. In: Current Practices and Controversies in Assisted Reproduction. Ed. by E. Vayena, P.J. Rowe et al. Geneva: World Health Organization, 2002: 152–65.
  155. Keel B.A., Webster B.W. Semen analysis data from fresh and cryopreserved donor ejaculates: comparison of cryoprotectants and pregnancy rates. Fertil. Steril. 1989; 52(1): 100–5.
  156. Watson P.F. Recent developments and concepts in the cryopreservation of spermatozoa and the assessment of their post-thawing function. Reprod. Fertil. Dev. 1995; 7(4): 871–91.
  157. Bagchi A., Woods E.J., Critser J.K. Cryopreservation and vitrification: recent advances in fertility preservation technologies. Expert. Rev. Med. Devices 2008; 5(3): 359–70.
  158. Sherman G.K. Cryopreservation of human semen. In: Handbook of Laboratory Diagnosis and Treatment of Infertility, 1st ed. Ed. by D.F. Keel, B.W. Webster. CRC-Press, 1990: 229–59.
  159. Vutyavanich T., Piromlertamorn W., Nunta S. Rapid freezing versus slow programmable freezing of human spermatozoa. Fertil. Steril. 2010; 93(6): 1921–8.
  160. Zribi N., Feki C.N., El E.H., Gargouri J., Bahloul A. Effects of cryopreservation on human sperm deoxyribonucleic acid integrity. Fertil. Steril. 2010; 93(1): 159–66.
  161. O’Flaherty C., Hales B.F., Chan P., Robaire B. Impact of chemotherapeutics and advanced testicular cancer or Hodgkin lymphoma on sperm deoxyribonucleic acid integrity. Fertil. Steril. 2010; 94(4): 1374–9.
  162. Woods E.J., Benson J.D., Agca Y., Critser J.K. Fundamental cryobiology of reproductive cells and tissues. Cryobiology 2004; 48(2): 146–56.
  163. Shin D., Lo K.C., Lipshultz L.I. Treatment options for the infertile male with cancer. J. Natl. Cancer Inst. Monogr. 2005; 34: 48–50.
  164. Meistrich M.L. Male gonadal toxicity. Pediatr. Blood Cancer 2009; 53(2): 261–6. 163. Feldschuh J., Brassel J., Durso N., Levine A. Successful sperm storage for 28 years. Fertil. Steril. 2005; 84(4): 1017.
  165. Horne G., Atkinson A.D., Pease E.H., Logue J.P., Brison D.R. Live birth with sperm cryopreserved for 21 years prior to cancer treatment: case report. Hum. Reprod. 2004; 19(6): 1448–9.
  166. Clarke G.N., Liu D.Y., Baker H.W. Recovery of human sperm motility and ability to interact with the human zona pellucida after more than 28 years of storage in liquid nitrogen. Fertil. Steril. 2006; 86(3): 721–2.
  167. Agarwal A., Ranganathan P., Kattal N. et al. Fertility after cancer: a prospective review of assisted reproductive outcome with banked semen specimens. Fertil. Steril. 2004; 81(2): 342–8.
  168. Saito K., Suzuki K., Iwasaki A., Yumura Y., Kubota Y. Sperm cryopreservation before cancer chemotherapy helps in the emotional battle against cancer. Cancer 2005; 104(3): 521–4.
  169. Schover L.R., Brey K., Lichtin A., Lipshultz L.I., Jeha S. Knowledge and experience regarding cancer, infertility, and sperm banking in younger male survivors. J. Clin. Oncol. 2002; 20(7): 1880–9.
  170. Janssens P.M., Beerendonk C.C., Blokzijl E., Braat D.D., Westphal J.R. Cryopreservation of semen of adolescents and young adult men with cancer. Ned. Tijdschr. Geneeskd. 2004; 148(40): 1981–4.
  171. Brinster R.L., Nagano M. Spermatogonial stem cell transplantation, cryopreservation and culture. Semin. Cell Dev. Biol. 1998; 9(4): 401–9.
  172. Nagano M., Patrizio P., Brinster R.L. Long-term survival of human spermatogonial stem cells in mouse testes. Fertil. Steril. 2002; 78(6): 1225–33.
  173. Avarbock M.R., Brinster C.J., Brinster R.L. Reconstitution of spermatogenesis from frozen spermatogonial stem cells. Nat. Med. 1996; 2(6): 693–6.
  174. Brook P.F., Radford J.A., Shalet S.M., Joyce A.D., Gosden R.G. Isolation of germ cells from human testicular tissue for low temperature storage and autotransplantation. Fertil. Steril. 2001; 75(2): 269–74.
  175. Keros V., Rosenlund B., Hultenby K., Aghajanova L., Levkov L. Optimizing cryopreservation of human testicular tissue: comparison of protocols with glycerol, propanediol and dimethylsulphoxide as cryoprotectants. Hum. Reprod. 2005; 20(6): 1676–87.
  176. Kvist K., Thorup J., Byskov A.G., Hoyer P.E., Mollgard K. Cryopreservation of intact testicular tissue from boys with cryptorchidism. Hum. Reprod. 2006; 21(2): 484–91.
  177. Radford J., Shalet S., Lieberman B. Fertility after treatment for cancer. Questions remain over ways of preserving ovarian and testicular tissue. BMJ 1999; 319(7215): 935–6.
  178. Radford J. Restoration of fertility after treatment for cancer. Horm. Res. 2003; 59(Suppl. 1): 21–3.
  179. Schlatt S., Rosiepen G., Weinbauer G.F., Rolf C., Nieschlag E. Germ cell transfer into rat, bovine, monkey and human testes. Hum. Reprod. 1999; 14(1): 144–50.
  180. Toyooka Y., Tsunekawa N., Akasu R., Noce T. Embryonic stem cells can form germ cells in vitro. Proc. Natl. Acad. Sci. USA 2003; 100(20): 11457–62.
  181. Yang S., Bo J., Hu H. et al. Derivation of male germ cells from induced pluripotent stem cells in vitro and in reconstituted seminiferous tubules. Cell Prolif. 2012; 45(2): 91–100.
  182. Honaramooz A., Snedaker A., Boiani M., Dobrinski I., Schlatt S. Sperm from neonatal mammalian testes grafted in mice. Nature 2002; 418(6899): 778–81.
  183. Honaramooz A., Li M.W., Penedo M.C., Meyers S., Dobrinski I. Accelerated maturation of primate testis by xenografting into mice. Biol. Reprod. 2004; 70(5): 1500–3.
  184. Ma P., Ge Y., Wang S., Ma J., Xue S. Spermatogenesis following syngeneic testicular transplantation in Balb/c mice. Reproduction 2004; 128(2): 163–70.
  185. Oatley J.M., de Avila D.M., Reeves J.J., McLean D.J. Spermatogenesis and germ cell transgene expression in xenografted bovine testicular tissue. Biol. Reprod. 2004; 71(2): 494–501.
  186. Schlatt S., Kim S.S., Gosden R. Spermatogenesis and steroidogenesis in mouse, hamster and monkey testicular tissue after cryopreservation and heterotopic grafting to castrated hosts. Reproduction 2002; 124(3): 339–46.
  187. Schlatt S., Honaramooz A., Boiani M., Scholer H.R., Dobrinski I. Progeny from sperm obtained after ectopic grafting of neonatal mouse testes. Biol. Reprod. 2003; 68(6): 2331–5.
  188. Shinohara T., Inoue K., Ogonuki N. et al. Birth of offspring following transplantation of cryopreserved immature testicular pieces and in-vitro microinsemination. Hum. Reprod. 2002; 17(12): 3039–45.
  189. Shinohara T., Orwig K.E., Avarbock M.R., Brinster R.L. Remodeling of the postnatal mouse testis is accompanied by dramatic changes in stem cell number and niche accessibility. Proc. Natl. Acad. Sci. USA 2001; 98(11): 6186–91.
  190. Ryu B.Y., Orwig K.E., Avarbock M.R., Brinster R.L. Stem cell and niche development in the postnatal rat testis. Dev. Biol. 2003; 263(2): 253–63.
  191. Jahnukainen K., Hou M., Petersen C., Setchell B., Soder O. Intratesticular transplantation of testicular cells from leukemic rats causes transmission of leukemia. Cancer Res. 2001; 61(2): 706–10.
  192. Gui Y.L., He C.H., Amory J.K. et al. Male hormonal contraception: suppression of spermatogenesis by injectable testosterone undecanoate alone or with levonorgestrel implants in Chinese men. J. Androl. 2004; 25(5): 720–7.
  193. Nieschlag E., Vorona E., Wenk M., Hemker A.K., Kamischke A. Hormonal male contraception in men with normal and subnormal semen parameters. Int. J. Androl. 2011; 34(6 Pt. 1): 556–67.
  194. Meistrich M.L., Zhang Z., Porter K.L. Prevention of adverse effects of cancer treatment on the germline. In: Male-mediated developmental toxicity. Ed. by D. Anderson, M.H. Brinkworth. Royal Society of Chemistry, 2007: 114–23.
  195. Dobrinski I., Ogawa T., Avarbock M.R., Brinster R.L. Effect of the GnRHagonist leuprolide on colonization of recipient testes by donor spermatogonial stem cells after transplantation in mice. Tissue Cell 2001; 33(2): 200–7.
  196. Meistrich M.L., Shetty G. Inhibition of spermatogonial differentiation by testosterone. J. Androl. 2003; 24(2): 135–48.
  197. Meistrich M.L., Shetty G. Hormonal suppression for fertility preservation in males and females. Reproduction 2008; 136(6): 691–701.
  198. Shetty G., Meistrich M.L. Hormonal approaches to preservation and restoration of male fertility after cancer treatment. J. Natl. Cancer Inst. Monogr. 2005; 34: 36–9.
  199. Howell S.J., Shalet S.M. Fertility preservation and management of gonadal failure associated with lymphoma therapy. Curr. Oncol. Rep. 2002; 4(5): 443–52.
  200. Meseguer M., Garrido N., Remohi J. et al. Testicular sperm extraction (TESE) and ICSI in patients with permanent azoospermia after chemotherapy. Hum. Reprod. 2003; 18(6): 1281–5.
  201. Binsaleh S., Sircar K., Chan P.T. Feasibility of simultaneous testicular microdissection for sperm retrieval and ipsilateral testicular tumor resection in azoospermic men. J. Androl. 2004; 25(6): 867–71.
  202. Baniel J., Sella A. Sperm extraction at orchiectomy for testis cancer. Fertil. Steril. 2001; 75(2): 260–2.
  203. Schrader M., Muller M., Sofikitis N. et al. “Onco-tese”: testicular sperm extraction in azoospermic cancer patients before chemotherapy-new guidelines? Urology 2003; 61(2): 421–5.
  204. Chan P.T., Palermo G.D., Veeck L.L., Rosenwaks Z., Schlegel P.N. Testicular sperm extraction combined with intracytoplasmic sperm injection in the treatment of men with persistent azoospermia postchemotherapy. Cancer 2001; 92(6): 1632–7.
  205. Damani M.N., Master V., Meng M.V. et al. Postchemotherapy ejaculatory azoospermia: fatherhood with sperm from testis tissue with intracytoplasmic sperm injection. J. Clin. Oncol. 2002; 20(4): 930–6.
  206. Zorn B., Virant-Klun I., Stanovnik M., Drobnic S., Meden-Vrtovec H. Intracytoplasmic sperm injection by testicular sperm in patients with aspermia or azoospermia after cancer treatment. Int. J. Androl. 2006; 29(5): 521–7.
  207. de Rooij D.G., van de Kant H.J., Dol R. et al. Long-term effects of irradiation before adulthood on reproductive function in the male rhesus monkey. Biol. Reprod. 2002; 66(2): 486–94.
  208. Hibi H., Ohori T., Yamada Y., Honda N., Hashiba Y. Testicular sperm extraction and ICSI in patients with post-chemotherapy non-obstructive azoospermia. Arch. Androl. 2007; 53(2): 63–5.
  209. Wyrobek A.J., Gordon L.A., Burkhart J.G. et al. An evaluation of human sperm as indicators of chemically induced alterations of spermatogenic function. A report of the U.S. Environmental Protection Agency Gene-Tox Program. Mutat. Res. 1983; 115(1): 73–148.
  210. Robbins W.A., Meistrich M.L., Moore D. et al. Chemotherapy induces transient sex chromosomal and autosomal aneuploidy in human sperm. Nat. Genet. 1997; 16: 74–8.
  211. Frias S., Van H.P., Meistrich M.L. et al. NOVP chemotherapy for Hodgkin’s disease transiently induces sperm aneuploidies associated with the major clinical aneuploidy syndromes involving chromosomes X, Y, 18, and 21. Cancer Res. 2003; 63(1): 44–51.
  212. Meistrich M.L. Effects of chemotherapy and radiotherapy on spermatogenesis. Eur. Urol. 1993; 23(1): 136–41.
  213. Wyrobek A.J., Schmid T.E., Marchetti F. Relative susceptibilities of male germ cells to genetic defects induced by cancer chemotherapies. J. Natl. Cancer Inst. Monogr. 2005; 34: 31–5.
  214. Chatterjee R., Haines G.A., Perera D.M., Goldstone A., Morris I.D. Testicular and sperm DNA damage after treatment with fludarabine for chronic lymphocytic leukaemia. Hum. Reprod. 2000; 15(4): 762–6.
  215. Meistrich M.L. Potential genetic risks of using semen collected during chemotherapy. Hum. Reprod. 1993; 8(1): 8–10. 215. Stahl O., Eberhard J., Jepson K. et al. Sperm DNA integrity in testicular cancer patients. Hum. Reprod. 2006; 21(12): 3199–205.
  216. Spermon J.R., Ramos L., Wetzels A.M. et al. Sperm integrity pre- and post-chemotherapy in men with testicular germ cell cancer. Hum. Reprod. 2006; 21(7): 1781–6.
  217. Dubrova Y.E., Grant G., Chumak A.A., Stezhka V.A., Karakasian A.N. Elevated minisatellite mutation rate in the post-Chernobyl families from Ukraine. Am. J. Hum. Genet. 2002; 71(4): 801–9.
  218. Brandriff B.F., Meistrich M.L., Gordon L.A., Carrano A.V., Liang J.C. Chromosomal damage in sperm of patients surviving Hodgkin’s disease following MOPP (nitrogen mustard, vincristine, procarbazine, and prednisone) therapy with and without radiotherapy. Hum. Genet. 1994; 93(3): 295–9.
  219. Tempest H.G., Ko E., Chan P., Robaire B., Rademaker A. Sperm aneuploidy frequencies analysed before and after chemotherapy in testicular cancer and Hodgkin’s lymphoma patients. Hum. Reprod. 2008; 23(2): 251–8.
  220. De M.P., Daudin M., Vincent M.C. et al. Increased aneuploidy in spermatozoa from testicular tumour patients after chemotherapy with cisplatin, etoposide and bleomycin. Hum. Reprod. 2001; 16(6): 1204–8.
  221. Winther J.F., Boice J.D. Jr., Mulvihill J.J. et al. Chromosomal abnormalities among offspring of childhood-cancer survivors in Denmark: a populationbased study. Am. J. Hum. Genet. 2004; 74(6): 1282–5.
  222. Blatt J. Pregnancy outcome in long-term survivors of childhood cancer. Med. Pediatr. Oncol. 1999; 33(1): 29–33.
  223. Byrne J., Rasmussen S.A., Steinhorn S.C. et al. Genetic disease in offspring of long-term survivors of childhood and adolescent cancer. Am. J. Hum. Genet. 1998; 62(1): 45–52.
  224. Dodds L., Marrett L.D., Tomkins D.J., Green B., Sherman G. Casecontrol study of congenital anomalies in children of cancer patients. BMJ 1993; 307(6897): 164–8.
  225. Green D.M., Whitton J.A., Stovall M. et al. Pregnancy outcome of partners of male survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J. Clin. Oncol. 2003; 21(4): 716–21.
  226. Kenney L.B., Nicholson H.S., Brasseux C. et al. Birth defects in offspring of adult survivors of childhood acute lymphoblastic leukemia. A Childrens Cancer Group/National Institutes of Health Report. Cancer 1996; 78(1): 169–76.
  227. Meistrich M.L., Byrne J. Genetic disease in offspring of long-term survivors of childhood and adolescent cancer treated with potentially mutagenic therapies. Am. J. Hum. Genet. 2002; 70(4): 1069–71.
  228. Senturia Y.D., Peckham C.S. Children fathered by men treated with chemotherapy for testicular cancer. Eur. J. Cancer 1990; 26(4): 429–32.
  229. Signorello L.B., Mulvihill J.J., Green D.M. et al. Congenital anomalies in the children of cancer survivors: a report from the childhood cancer survivor study. J. Clin. Oncol. 2012; 30(3): 239–45.
  230. Tiemann-Boege I., Navidi W., Grewal R. et al. The observed human sperm mutation frequency cannot explain the achondroplasia paternal age effect. Proc. Natl. Acad. Sci. USA 2002; 99(23): 14952–7.
  231. Glaser R.L., Broman K.W., Schulman R.L. et al. The paternal-age effect in Apert syndrome is due, in part, to the increased frequency of mutations in sperm. Am. J. Hum. Genet. 2003; 73(4): 939–47.
  232. Giwercman A. Gonadotoxic cancer treatment in males—a reason for andrological counselling? Radiother. Oncol. 2003; 68(3): 213–5.
  233. Pierik F.H., Dohle G.R., van Muiswinkel J.M., Vreeburg J.T., Weber R.F. Is routine scrotal ultrasound advantageous in infertile men? J. Urol. 1999; 162(5): 1618–20.
  234. Pierik F.H., Van Ginneken A.M., Dohle G.R., Vreeburg J.T., Weber R.F. The advantages of standardized evaluation of male infertility. Int. J. Androl. 2000; 23(6): 340–6.
  235. Wang J., Galil K.A., Setchell B.P. Changes in testicular blood flow and testosterone production during aspermatogenesis after irradiation. J. Endocrinol. 1983; 98(1): 35–46.
  236. Setchell B.P., Galil K.A. Limitations imposed by testicular blood flow on the function of Leydig cells in rats in vivo. Aust. J. Biol. Sci. 1983; 36(3): 285–93.

Hodgkin’s lymphoma and a “new old” bendamustine

S.S. Shklyaev, and V.V. Pavlov

Medical Radiological Research Center, RF Ministry of Health, Obninsk, Russian Federation


ABSTRACT

Hodgkin’s lymphoma is a malignant tumor that eventually turned from a fatal incurable to successfully curable disease after primary treatment in the vast majority of cases. However, the prognosis for patients with refractory and relapsed disease is not infrequently dismal and life-threatening, especially if the tumor continues progressing after high-dose chemotherapy with autologous stem cell transplantation or, in some instances, even after allogeneic stem cell grafting. Bendamustine is a “new old” cytostatic agent that can be effectively applied for treating this group of patients. Our literature review highlights a variety of relevant options in treatment of Hodgkin’s lymphoma using bendamustine.


Keywords: Hodgkin’s lymphoma, refractory and relapsed disease, treatment, bendamustine.

Read in PDF (RUS)pdficon


REFERENCES

  1. Hodgkin T. On some morbid appearances of the absorbent glands and spleen. Med. Chir. Trans. 1832; 17: 68–114.
  2. Wilks S. Cases of enlargement of the lymphatic glands and spleen (or Hodgkin’s disease), with remarks. Guy’s Hosp. Rep. 1865; 11: 56–67.
  3. Sternberg K. Uber eine eigenartige unter dem Bilde der Pseudoleukamie verlaufende Tuberculose des lymphatischen Apparates. Z. Heilk. 1898; 19: 21–90.
  4. Rosenfield L. Hodgkin’s disease: origin of an eponym — and one that got away. Bull. N. Y. Acad. Med. 1989; 65: 618–32.
  5. Winiwarter A. Arch. Klin. Chir. 1875; 18: 98–102.
  6. Trousseau A. De l’adenie. Clin. Med. De Hotel-Dieu 1877; III: 609.
  7. Pusey W.A. Cases of sarcoma and of Hodgkin’s disease treated by exposure to X-rays: a preliminary report. J. Am. Med. Assoc. 1902; 38: 166–9.
  8. Goodman L.S., Wintrobe M.M., Dameshek W. et al. Nitrogen mustard therapy; use of methyl-bis (beta-chloroethyl) amine hydrochloride and tris (beta-chloroethyl) amine hydrochloride for Hodgkin’s disease, lymphosarcoma, leukemia and certain allied and miscellaneous disorders. J. Am. Med. Assoc. 1946; 132: 126–32.
  9. De Vita V., Serpick A. Combination chemotherapy in the treatment of advanced Hodgkin’s disease. Proc. Am. Assoc. Cancer Res. 1967; 8: 13.
  10. De Vita V., Serpick A., Carbone P.P. Combination chemotherapy in the treatment of advanced Hodgkin’s disease. Ann. Int. Med. 1970; 73: 891–5.
  11. Fairley G.H., Patterson M.J., Scott R.B. Chemotherapy of Hodgkin’s Disease with Cyclophosphamide, Vinblastine, and Procarbazine. Br. Med J. 1966; 2(5505): 75–8.
  12. Bonadonna G., Zucali R., Monfardini S. et al. Combination chemotherapy of Hodgkin’s disease with adriamycin, bleomycin, vinblastine, and imidazole carboxamide versus MOPP. Cancer 1975; 36(1): 252–9.
  13. Canellos G.P., Anderson J.R., Propert K.J. et al. Chemotherapy of advanced Hodgkin’s disease with MOPP, ABVD, or MOPP alternating with ABVD. N. Engl. J. Med. 1992; 327(21): 1478–84.
  14. Diehl V. Dose-escalation study for the treatment of Hodgkin’s disease. The German Hodgkin Study Group (GHSG). Ann. Hematol. 1993; 66(3): 139–40.
  15. Connors J.M. State-of-the-art therapeutics: Hodgkin’s lymphoma. J. Clin. Oncol. 2005; 23(26): 6400–8.
  16. Brenner H., Gondos A., Pulte D. Ongoing improvement in long-term survival of patients with Hodgkin disease at all ages and recent catch up of older patients. Blood 2008; 111: 2977–83.
  17. Павлов В.В., Богатырева Т.И., Шахтарина С.В., Даниленко А.А. Оптимизация лучевого компонента в программах комбинированного химио лучевого лечения больных лимфомой Ходжкина. В кн.: Терапевти- ческая радиология. Руководство для врачей. Под ред. А.Ф. Цыба, Ю.С. Мардынского. М.: ООО «МК», 2010: 461–505. Pavlov V.V., [Bogatyreva T.I., Shakhtarina S.V., Danilenko A.A. Optimizatsiya luchevogo komponenta v programmakh kombinirovannogo himioluchevogo lecheniya bolnikh limfomoy Hodzhkina. V kn.: Terapevticheskaya radiologiya. Rukovodstvo dlya vrachey. Pod red. A.F. Tsyba, Yu.S. Mardynskogo [Optimization of radiotherapy component in combined chemoradiotherapy programs for patients with Hodgkin’s lymphoma. In: Therapeutic radiology. Manual for medical practitioners. Ed. by A.F. Tsyba, Yu.S. Mardynsky. M.: OOO “MK”, 2010: 461–505.]
  18. Ansell S.M., Armitage J.O. Management of Hodgkin lymphoma. Mayo Clin. Proc. 2006; 81(3): 419–26.
  19. Richardson S.E., McNamara C. The Management of Classical Hodgkin’s Lymphoma: Past, Present, and Future. Adv. Hematol. 2011; 2011: 865870. doi: 10.1155/2011/865870.
  20. Holmberg L., Maloney D.G. The role of autologous and allogeneic hematopoietic stem cell transplantation for Hodgkin lymphoma. J. Natl. Compr. Canc. Netw. 2011; 9(9): 1060–71.
  21. Богатырева Т.И., Павлов В.В., Шкляев С.С. Рецидивы лимфомы Ход- жкина: возможности продления жизни без высокодозной химиотерапии. Врач 2012; 11: 5–8.  [Bogatyreva T.I., Pavlov V.V., Sklyayev S.S. Retsidivy limfomy Hodzhkina: vozmozhnosti prodleniya zhizni bez vysokodoznoy himioterapii [Hodgkin’s lymphoma relapse: possibilities of life prolongation without high-dose chemotherapy. Vrach 2010; 11: 5–8.]
  1. Павлов В.В. Кроветворение при дистанционной гамма-терапии в условиях ежедневного ритма облучения: Автореф. дис. ¼ канд. мед. наук. М., 1973. Pavlov V.V. [Krovetvoreniye pri distantsionnoy gamma-terapii v usloviyakh ezhednevnogo ritma oblucheniya: Avtoref. dis. … kand. med. nauk [Hemopoiesis during X-ray therapy using daily rhythm irradiation. Author’s summary of dissertation for the degree of Candidate of medical sciences. M., 1973.]
  2. Younes A., Gopal A.K., Smith S.E. et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J. Clin. Oncol. 2012; 30(18): 2183–9.
  3. Goyal S.D., Bartlett N.L. Where does brentuximab vedotin fit into the management of patients with Hodgkin lymphoma? Curr. Hematol. Malig. Rep. 2012; 7(3): 179–85.
  4. de Claro RA, McGinn K, Kwitkowski V et al. U.S. Food and drug administration approval summary: brentuximab vedotin for the treatment of relapsed Hodgkin lymphoma or relapsed systemic anaplastic large-cell lymphoma. Clin. Cancer Res. 2012; 18(21): 5845–9.
  5. Moskowitz A.J. Novel agents in Hodgkin lymphoma. Curr. Oncol. Rep. 2012; 14(5): 419–23. doi: 10.1007/s11912-012-0251-y. Review.
  6. Canellos G.P. Brentuximab vedotin and panobinostat; new drugs for Hodgkin’s lymphoma — can they make one of medical oncology’s chemotherapy success stories more successful? J. Clin. Oncol. 2012; 30(18): 2171–2. doi: 10.1200/JCO.2011.39.6416.
  7. Lemoine M., Derenzini E., Buglio D. et al. The pan-deacetylase inhibitor panobinostat induces cell death and synergizes with everolimus in Hodgkin. Blood 2012; 119(17): 4017–25. doi: 10.1182/blood-2011- 01-331421.
  8. Guarini A., Minoia C., Giannoccaro M. et al. mTOR as a target of everolimus in refractory/relapsed Hodgkin lymphoma. Curr. Med. Chem. 2012; 19(7): 945–54.
  9. Mandac I., Kolonic S.O. Lenalidomide induced good clinical response in a patient with multiple relapsed and refractory Hodgkin’s lymphoma. J. Hematol. Oncol. 2010; 3: 20. doi: 10.1186/1756-8722-3-20.
  10. Kalaycio M. Bendamustine: a new look at an old drug. Cancer 2009; 115: 473–9.
  11. Ozegowski W., Krebs D. w-[bis-(chlorethyl)-amino-benzimidazolyl-(2)]- propionic or butyric acids as potential cytostatic agents. J. Prakt. Chem. 1963; 20: 178–86.
  12. Ozegowski W., Krebs D. IMET 3393, gamma-(1-methyl-5-bis-(b-chlrathyl)- amino-benzimidazlolyl(2)-buttersaure-hydrochlorid, ein neues Zytostatikum aus der Reihe der Benzimidazol-Loste. Zbl. Pharm. 1971; 110: 1013–9.
  13. Anger G., Hesse P., Kohler P., Baufeld H. Erste klinische Erfahrungen mit einem neuen Zytostatikum. Deutsch Gesundheitswes. 1967; 22: 1079–84.
  14. Anger G., Hesse P., Baufeld H. Behandlung des multiplen Myeloms mit einem neuen Zytostatikum. Dtsch. Med. Wochenschr. 1969; 48: 2495–500.
  15. Anger G., Fink R., Fleischer J. et al. Vergleichsuntersuchungen zwischen Cytostasan und Cyclophosphamid bei der chronischen Lymphadenose, dem Plasmozytom, der Lymphogranulomatose und dem Bronchialkarzinom. Dt. Gesundh. Wesen. 1975; 30: 1280–5.
  16. Schnabel R., Jungstand W., Gutsche W. et al. Comparative studies on the cytostatic activity of the nitrogen mustard derivative IMET 3393 and endoxan in three experimental mouse tumors (Ehrlich-ascites-carcinoma, sarcoma 180 solid, leukaemia LAJ 1). Acta Biol. Med. 1967; 19: 534–58.
  17. Hoche D., Wutke K., Anger G. et al. Vergleichende Untersuchung zur Wirksamkeit des DBVCy-Protocolls mit dem ABVD-Protokoll beim fortgeschrittenen Hodgkin Lymphom. Arch. Geschwulstforsch. 1984; 54(4): 333–42.
  18. Herold M., Anger G., Hoche D., Kastner R. Vorlaufige Ergebnisse einer zyklisch-alternierenden Chemotherapie (CVPP/DBVCy) bei fortgeschrittenem Morbus Hodgkin. Med. Klin. 1987; 82(10): 345–9.
  19. Dennie T.W., Kolesar J.M. Bendamustine for the treatment of chronic lymphocytic leukemia and rituximab-refractory, indolent B-cell non-Hodgkin lymphoma. Clin. Ther. 2009; 31: 2290–311.
  20. Cheson B.D., Wendtner C.M., Pieper A. et al. Optimal use of bendamustine in chronic lymphocytic leukemia, non-Hodgkin lymphomas, and multiple myeloma: treatment recommendations from an international consensus panel. Clin. Lymphoma Myeloma Leuk. 2010; 10(1): 21–7.
  21. Borchmann P., Schnell R., Diehl V., Engert A. New drugs in the treatment of Hodgkin’s disease. Ann. Oncol. 1998; Suppl. 5: S103–8. 43. http://ash.confex.com/ash/2009/webprogram/Paper16834.html.
  22. Moskowitz A.J., Hamlin P.A., Gerecitano J. et al. Bendamustine is highly active in heavily pre-treated relapsed and refractory Hodgin’s lymphoma and serves as a bridge to allogeneic stem cell transplant. Blood 2009; 114: Abstract 720.
  23. Moskowitz A., Perales M., Kewalramani T. et al. Outcomes for patients who fail high dose chemoradiotherapy and autologous stem cell rescue for relapsed and primary refractory Hodgkin lymphoma. Br. J. Haematol. 2009; 146: 158–63.
  24. http://clinicaltrials.gov/ct2/show/NCT00705250?term=Bendamustine+in+ Relapsed+and+Primary+Refractory+Hodgkin%27s+Lymphoma&rank=2
  25. De Flippi R., Aldinucci D., Galati D. et al. Effect of bendamustine on apoptosis and colony-initiating precursors in Hodgkin lymphoma cells. J. Clin. Oncol. 2011; 29(Suppl.): Abstract e18559.
  26. Leoni L.M., Bailey B., Reifert J. et al. Bendamustine (Treanda) displays a distinct pattern of cytotoxicity and unique mechanistic features compared with other alkylating agents. Clin. Cancer Res. 2008; 14(1): 309–17.
  27. Leoni L.M., Niemeyer C.C., Kerfoot C. et al. In vitro and ex vivo activity of SDX-105 (bendamustine) in drug-resistant lymphoma cells [abstract 1215]. Proc. Am. Assoc. Cancer Res. 2004; 45: 278.
  28. Friedberg J.W., Cohen P., Chen L. et al. Bendamustine in patients with rituximab-refractory indolent and transformed non-Hodgkin’s lymphoma: results from a phase II multicentere, single-agent study. J. Clin. Oncol. 2008; 26(2): 204–10.
  29. Konstantinov S.M., Kostovski A., Topashka-Ancheva M. et al. Cytotoxic efficacy of bendamustine in human leukemia and breast cancer cell lines. J. Cancer Res. Clin. Oncol. 2002; 128(5): 271–8.
  30. Leoni L.M., Hartley J.A. Mechanism of action: the unique pattern of bendamustine-induced cytotoxicity. Semin. Hematol. 2011; 48(Suppl. 1): S12–23.
  31. Furukawa Y., Hiraoka N., Wada T. et al. Mechanisms of action and clinical effectiveness of the newly approved anti-cancer drug bendamustine. Nihon Yakurigaku Zasshi (Folia Pharmacol. Jpn.). 2011; 138(1): 26–32.
  32. Beeharry N., Rattner J.B., Belacosa A. et al. Dose dependent effects on cell cycle checkpoints and DNA repair by bendamustine. PLoS One. 2012; 7(6): e40342.
  33. Стругов В.В., Стадник Е.А., Зарицкий А.Ю. Механизм действия и кли- ническая эффективность нового алкилирующего препарата бендамустин при хроническом лимфолейкозе. Клин. онкогематол. 2011; 4(3): 217–27. [Strugov V.V., Stadnik E.A., Zaritskiy A.Yu. Mekhanizm deystviya i klinicheskaya effectivnost novogo alkiliruyushchego preparata bendamustin pri khronicheskom limfoleykoze [Mechanism of action and clinical efficacy of bendamustine, a new alkylating agent, in chronic lymphocytic leukemia]. Klin. onkogematol. 2011; 4(3): 217–27.]
  1. D’Elia G.M., De Anelis F., Breccia M. et al. Efficacy of bendamustine as salvage treatment in an heavily pre-treated Hodgkin lymphoma. Leuk. Res. 2010; 34(11): e300–1.
  2. Magyari F., Simon Z., Barna S. et al. Successful administration of rituximab-bendamustine regimen in the relapse of Hodgkin lymphoma after autologous hemopoietic stem cell transplantation. Hematol. Oncol. 2012; 30(2): 98–100.
  3. Jones R.J., Gocke C.D., Kasamon Y.L. et al. Circulating clonotypic B cells in classic Hodgkin lymphoma. Blood 2009; 113(23): 5920–6.
  4. Younes A., Oki Y., McLaughlin P. et al. Phase 2 study of rituximab plus ABVD in patients with newly diagnosed classical Hodgkin lymphoma. Blood 2012; 119(18): 4123–8.
  5. Kasamon Y.L., Jacene H.A., Gocke C.D. et al. Phase 2 study of rituximabABVD in classical Hodgkin lymphoma. Blood 2012; 119(18): 4129–32.
  6. Rummel M.J., Chow K.U., Hoelzer D. et al. In vitro studies with bendamustine: enhanced activity in combination with rituximab. Semin. Oncol. 2002; 29(4 Suppl. 13): 12–4.
  7. Currin E.S., Gopal A.K. Treatment strategies for Hodgkin lymphoma recurring following autologous hematopoietic stem cell transplantation. Korean J. Hematol. 2012; 47(1): 8–16.
  8. Mian M., Farsad M., Pescosta N. et al. Bendamustine salvage for the treatment of relapsed Hodgkin’s lymphoma after allogeneic bone marrow transplantation. Ann. Hematol. 2012; published online 22.07.2012.
  9. Corazzelli G., Angrilli F., D’Arco A. et al. Efficacy and safety of bendamustine for the treatment of patients with recurring Hodgkin lymphoma. Br. J. Haematol. 2013; 160(2): 207–15. doi: 10.1111/bjh.12120. [Epub ahead of print 2012 Nov. 20].
  10. Visani G., Malerba L., Stefani P.M. et al. BeEAM (bendamustine, etoposide, cytarabine, melphalan) before autologous stem cell transplantation is safe and effective for resistant/relapsed lymphoma patients. Blood 2011; 118(12): 3419–25.
  11. Регистрация клинического исследования в Европейском медицинском агентстве (European Medicines Agency): EudraCT #2008-002736-15. [Registratsiya klinicheskogo issledovaniya v Evropeyskom meditsinskom agentstve (European Medicines Agency) [Registration of a clinical study in European Medicines Agency]. EudraCT #2008-002736-15.]
  1. Kath R., Blumenstengel K., Fricke H.J. et al. Bendamustine, vincristine; prednisolone (BOP) in therapy of advanced low-grade non-Hodgkin lymphoma. Dtsch. Med. Wochenschr. 2001; 126(8): 198–202.
  2. Friedberg J.W., Vose J.M., Kelly J.L. et al. The combination of bendamustine, bortezomib, and rituximab for patients with relapsed/refractory indolent and mantle cell non-Hodgkin lymphoma. Blood 2011; 117(10): 2807–12.
  3. Visco C., Castegnaro S., Chieregato K. et al. The cytotoxic effects of bendamustine in combination with cytarabine in mantle cell lymphoma cell lines. Blood Cells. Mol. Dis. 2012; 48(1): 68–75.
  4. Yong H.X., Linn Y.C., Ong K.H. et al. Chemoimmunotherapy with bendamustine hydrochloride and alemtuzumab demonstrates synergism in T-prolymphocytic leukemia. Leuk. Res. 2012; 36(8): e163–5.
  5. Alaikov T., Konstantinov S.M., Tzanova T. et al. Antineoplastic and anticlastogenic properties of curcumin. Ann. N. Y. Acad. Sci. 2007; 1095: 355–70.
  6. Lentzsch S., O’Sullivan A., Kennedy R.C. et al. Combination of bendamustine, lenalidomide, and dexamethasone (BLD) in patients with relapsed or refractory multiple myeloma is feasible and highly effective: results of phase 1/2 open-label, dose escalation study. Blood 2012; 119(20): 4608–13.
  7. Loibl S., Doering G., Muller L. et al. Phase II Study with Weekly Bendamustine and Paclitaxel as First- or Later-Line Therapy in Patients with Metastatic Breast Cancer: RiTa II Trial. Breast Care (Basel). 2011; 6(6): 457–61.
  8. Rahn A.N., Schilcher R.B., Adamietz I.A. et al. Palliative radiochemotherapy with Bendamustine for advanced recurrent head and neck tumors. Strahlenther. Onkol. 2001; 177(4): 189–94.
  9. Bottke D., Bathe K., Wiegel T., Hinkelbein W. Phase I trial of radiochemotherapy with bendamustine in patients with recurrent squamous cell carcinoma of the head and neck. Strahlenther. Onkol. 2007; 183(3): 128–32.
  10. Moskowitz A.J., Hamlin P.A. Jr., Perales M.A. et al. Phase II Study of Bendamustine in Relapsed and Refractory Hodgkin Lymphoma. J. Clin. Oncol. 2012 Dec 17 [Epub ahead of print].