Acessibilidade / Reportar erro

Fertility in female survivors of Hodgkin's lymphoma

Abstract

Currently, Hodgkin's lymphoma is one of the most curable types of cancer. Patients are often young and so the long-term morbidities of treatment have become of increasing concern. Among these, infertility is one of the most challenging consequences for patients in reproductive age. Premature ovarian failure in premenopausal women is a serious long-term sequel of the toxicity of chemotherapy. The main consequence of this syndrome is infertility, but women also present other symptoms related to estrogen deprivation. Different rates of impaired gonadal function are reported, depending on the patient's age, stage of disease, dose and intensity of chemotherapy and the use of radiation therapy. The most established strategy in female infertility is cryopreservation of embryos after in vitro fertilization. Additionally, the use of oral contraceptives or gonadotropinreleasing hormone analogs (GnRH-a) during treatment is under study. This review will provide a general overview of the main studies conducted to evaluate the infertility rate among female Hodgkin's lymphoma survivors and risk factors associated to treatment, different end-point definitions for evaluating fertility and also a brief description of the available strategies for fertility preservation.

Hodgkin's lymphoma; Fertility; Survivors


SPECIAL ARTICLE

Fertility in female survivors of Hodgkin's lymphoma

Irene BiasoliI; Simona FalorioII; Stefano LuminariIII; Nelson SpectorI; Massimo FedericoIII

IHematology and Pathology Services, Hospital Universitário, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil

IIDepartment of Hematology, Ospedale Civile Santo Spirito, Pescara, Italy

IIIDepartment of Oncology, Hematology and Respiratory Diseases, L'Università di Modena e Reggio Emilia - UniMoRe, Modena, Italy

Corresponding author Corresponding author: Massimo Federico Department of Oncology, Hematology and respiratory diseases University of Modena and Reggio Emilia Centro Oncologico Modenese Via del pozzo 71 41124 Modena, Italy Phone: 39 059 4225515 federico@unimore.it

ABSTRACT

Currently, Hodgkin's lymphoma is one of the most curable types of cancer. Patients are often young and so the long-term morbidities of treatment have become of increasing concern. Among these, infertility is one of the most challenging consequences for patients in reproductive age. Premature ovarian failure in premenopausal women is a serious long-term sequel of the toxicity of chemotherapy. The main consequence of this syndrome is infertility, but women also present other symptoms related to estrogen deprivation. Different rates of impaired gonadal function are reported, depending on the patient's age, stage of disease, dose and intensity of chemotherapy and the use of radiation therapy. The most established strategy in female infertility is cryopreservation of embryos after in vitro fertilization. Additionally, the use of oral contraceptives or gonadotropinreleasing hormone analogs (GnRH-a) during treatment is under study. This review will provide a general overview of the main studies conducted to evaluate the infertility rate among female Hodgkin's lymphoma survivors and risk factors associated to treatment, different end-point definitions for evaluating fertility and also a brief description of the available strategies for fertility preservation.

Keywords: Hodgkin's lymphoma; Fertility; Survivors

Introduction

Due to the large body of randomized trials conducted by cooperative groups in North America and Europe, the outcome of Hodgkin's lymphoma (HL) has greatly improved over recent decades and HL is today among the most curable malignancies. However, new strategies for further improving HL outcomes in patients should take into account two major concerns: first, a reduction in long-term treatment-related toxicity and second, a decrease in the failure rate for advanced stages.(1)

Among the complications, infertility is one of the most challenging consequences for patients in reproductive age. In female survivors, one of major concerns is the risk of premature ovarian failure (POF).(2) POF is defined as the premature termination of ovarian function of peripheral origin and is a serious long-term sequel of toxicity. The main consequence of this syndrome is infertility, but women also present symptoms related to estrogen deprivation, such as osteoporosis. Different rates of secondary amenorrhea are reported, depending on the patient's age, stage of disease, dose and intensity of chemotherapy and use of radiation therapy. The most established strategy in female infertility is the cryopreservation of embryos after in vitro fertilization. Also, the use of oral contraceptives or gonadotropin-releasing hormone analogs (GnRH-a) during treatment aimed at preventing secondary ovarian failure is under study.(3-5)

This review will provide a general overview of the main studies conducted to evaluate the frequency of infertility among female HL survivors and risk factors associated to treatment, different end-point definitions for evaluating fertility and also, a brief description of the methods of fertilization preservation.

General issues and definitions

The ovaries contain a pool of primordial follicles that decreases during life. At birth, there are about two million follicles, whereas 200 000 remain at puberty and 25 000 at the age of 37 years-old. At the time of menopause there is a pool of fewer than 1000 follicles, when the follicle-stimulating hormone (FSH) levels are insufficient due to declining estrogen levels.(6)

The number of remaining follicles in the ovaries is called the ovarian reserve and represents the woman's current reproductive capacity. The anti-Mullerian hormone (AMH) is produced by early follicles. It has an inhibitory effect on primordial follicle recruitment as well as on the responsiveness of growing follicles to FSH. Its level declines with age and becomes undetectable after menopause.

In premenopausal women, ovarian function is controlled by FSH and luteinizing hormone (LH) produced in the pituitary. FSH activates the granulosa cells of growing ovarian follicles, which proliferate and produce estradiol. This reduces the levels of FSH by feedback inhibition, thus keeping them at low levels. LH at the middle of the cycle induces ovulation following the formation of the luteal body that produces progesterone. The growing follicles also produce inhibin, which prevents an overgrowth of follicles by down regulating FSH.(7)

The main consequence of POF is infertility, but women may also present hot flushes, atrophic vaginitis and osteoporosis. Cancer treatment may induce acute ovarian failure (AOF) shortly after the conclusion of treatment.(8,9) A proportion of these women resume normal menses within months. Among women who maintain ovarian function after the end of treatment, some may present a premature menopause before the age of 40 years old. In addition to the impact on the quality of life, premature menopause may increase the risk of cardiovascular disease and osteoporosis.(9)

Infertility risk factors

Age

Age and the status of the ovarian reserve before treatment are risk factors for POF in HL. Some studies reported an increase in risk in over 30-year-old women, but this is probably an arbitrary cut-off. Age-related gonadal injury may also be influenced by a natural decline in fertility with increased age. Younger women tolerate higher cumulative doses of chemotherapy before developing amenorrhea and have a greater chance of resuming menses after treatment. On the other hand, older women, who have an already depleted number of follicles at the onset of treatment, are more susceptible to gonadal toxicity.(3,8,10)

Treatment

While it is well-established that combination chemotherapy causes depletion of the ovarian follicle reserve, the gonadotoxicity of each agent is not easy to establish. Alkylating agents are most often associated with dose-dependent and irreversible gonadal toxicity. The risk of POF is about 50% in young women receiving cyclophosphamide at a mean cumulative dose of 9.3 g. Moreover, there is an 11% increase in the risk of POF with each additional 1.4 g/m2 of cumulative procarbazine.(3,8,10) A summary of the risks of premature menopause of each drug is shown in Table 1.

Currently,ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine) and escalated BEACOPP (bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, prednisone) are the most frequently used regimens for treating HL.(1) In general, ABVD is not associated with a greater risk of premature menopause. Hodgson et al. reported the results of a case-control study of thirty-six female HL survivors who had attempted pregnancy and found no evidence of significant impairment in fertility compared to controls.(11) The German Hodgkin Study Group (GHSG) analyzed the menstrual status after HL treatment of 405 patients.(10) They found a proportion of 51.4% of the women who had received eight cycles of escalated BEACOPP had permanent amenorrhea. The escalated regimen produced a higher rate of amenorrhea than standard BEACOPP.

In a large cohort of 518 female HL survivors, after a median follow-up of 9.4 years, 97 women reached menopause before the age of 40 years.(8) Chemotherapy was associated with a 12.3-fold increase in premature menopause, when compared to radiotherapy alone. Also, abdominal or pelvic radiation increases the risk of POF.

The literature concerning the risks of infertility after hematopoietic cell transplantation (HCT) is scarce. Some studies have pointed out that the risk with allogeneic stemcell transplantation is the highest, especially after total body irradiation (TBI). However, a report from the Late Effects Working Committee of the Center for International Blood and Marrow Transplant Research (CIBMTR) revealed the occurrence of 83 pregnancies in female HCT recipients. The series included recipients of autologous HCT, myeloablative allogeneic HCT and non-myeloablative allogeneic HCT.(12)

A summary of the main studies about risk factors of impaired gonadal function in female HL patients is presented in Table 2.

Evaluation of fertility

Infertility is defined as the failure to conceive after one year of regular intercourses without taking contraceptive measures. Infertility secondary to cancer treatment may be transient, permanent or delayed.(5)

The diagnosis of POF is based on clinical and laboratory evidence. Ovarian reserve tests (ORT) include ultrasound and serum biomarkers.(4,8,17)

Clinical evidence is based on the presence or absence of menstrual cycles. After treatment, the recovery of normal menstrual cycles does not warrant normal fertility, but amenorrhea is the strongest predictor of infertility.Also, the presence of transient amenorrhea after treatment is a risk factor for subsequent infertility.(4,17)

Laboratory evidence includes determining the serum levels of FSH, AMH and inhibin B. Usually, levels of FSH are elevated in cases of impaired fertility, but normal levels do not exclude impaired ovarian function. The AMH is undetectable after menopause. Also, the level of inhibin, which is secreted by the follicles, decreases with age.

Moreover, transvaginal ultrasound has been evaluated in the diagnosis of POF. The antral follicle count (AFC) and also the ovarian volume are useful parameters associated with ovarian reserve and fertility.(18)

It is also known that patients with Hodgkin's lymphoma may face infertility at the time of diagnosis, that is, before treatment. Data come mostly from studies performed in men, but it is possible that similar mechanisms operate in women. Some studies evaluating semen quality before treatment showed that most patients had azoospermia or dyspermia. The underlying mechanism of infertility in HL patients is still unknown. Possible factors are damage to the germinal epithelium and disorders of the hypothalamic-hypophysial axis as well as the impact of cytokines on spermatogenesis.(19)

Fertility preservation strategies

Assisted reproduction

The most established strategy for female infertility is assisted reproduction (embryo and oocyte cryopreservation). However, this approach has some limitations. Only pubertal patients and patients with a long-term partner can benefit from this procedure. Also, patients presenting an urgent medical complication cannot wait for the delay imposed by the procedure, since the process usually requires a delay of one month before starting chemotherapy. Alternatives under study are fertilization of oocytes by intracytoplasmic sperm injection and oocyte vitrification.(4,20)

Cryopreservation of ovarian tissue

This represents an investigational method offered to pre-pubertal and pubertal females. The general principle is to remove the ovary or obtain an ovarian cortex biopsy and freeze the primordial follicles using established protocols. Nevertheless, only 12 full-term pregnancies using this method have been reported so far, and among them only 4 in patients with HL. So, the results are still uncertain.(4,21-23)

Pharmacological methods - Gonadotropin-releasing hormone analogues (GnRH) and oral contraceptives

There are many suggested mechanisms of protection against gonadotoxicity by GnRH-a.(24) The first is the creation of a hormonal milieu similar to the pre-pubertal state. It has been hypothesized that chemotherapy induces an accelerated rate of follicular destruction with subsequent decreases in the production of estradiol and inhibin resulting in increases in FSH production. This increase in FSH causes enhanced recruitment of follicles and subsequent destruction. The administration of GnRH or oral contraceptives may prevent the increased levels of FSH by inducing pituitary desensitization, thus protecting the follicles. Another possible mechanism is decreased uteroovarian perfusion. It has been speculated that this decrease in perfusion could reduce the exposure of ovaries to chemotherapeutic agents. Finally, a potential direct effect on GnRH receptors has been proposed.

Several studies have been published to establish the role of GnRH-a and oral contraceptives in preserving fertility in female cancer patients. However, the literature is still controversial.(24,25) In a retrospective study, GNRH-a allowed the recovery of menses in 92% of patients and all 13 patients that attempted pregnancy conceived.(3) In another prospective study, 97% of patients taking GnRH-a resumed ovulation and regular menses compared with 63% control subjects.(26) On the other hand, a randomized prospective trial aimed to compare oral contraceptives and GNRH-a in female HL patients treated with escalated BEACOPP was recently closed because neither arm protected ovarian function as measured by AMH levels.(27) Furthermore, the authors that are against the recommendation of the use of GnRH for young female patients outside clinical trials argue that: (i) most of the data came from retrospective studies with limited numbers of patients, (ii) there is a lack of prospective, randomized clinical trials, (iii) there is a lack of biological plausibility for protection by GnRH and also, (iv) there are concerns about the safety of the procedure.(25) On the other hand, some other authors consider that the published clinical studies provide some evidence that GnRH-a reduces gonadotoxicity and GnRH-a can even reduce some chemotherapy-induced complications, such as severe menometrorrhagia. In 2011, the results of a randomized trial, PROMISE-GIM6, showed that the use of triptorelin during chemotherapy in premenopausal patients with early-stage breast cancer reduced the occurrence of early chemotherapyinduced menopause. A summary of the main studies is presented in Table 3.

Impairment of reproductive function can be a significant late effect of cancer treatment, mainly for young female patients. Understanding the major risk factors and the role of alternative strategies such as the use of GnRH-a or oral contraceptives during treatment will help prevent gonadal damage. The benefits and risks of each treatment regimen must be discussed individually, mainly with female patients older than 30 years old and those receiving alkylating-containing regimens. Also, if available, patients should be offered assisted reproductive consultations. Finally, a careful assessment of the ovarian function before treatment should be performed in all circumstances and the collection of such information should be encouraged when planning prospective clinical trials as well as lymphoma registries.

Acknowledgments

Financial support provided by the Associazione Angela Serra per la Ricerca sul Cancro, Modena, Italy and FAPERJ (Fundação de Amparo à Pesquisa do Rio de Janeiro)

Submitted: 12/1/2011

Accepted: 12/4/2011

Conflict-of-interest disclosure: The authors declare no competing financial interest

www.rbhh.org or www.scielo.br/rbhh

  • 1. Borchmann P, Engert A. The past: what we have learned in the last decade. Hematology Am Soc Hematol Educ Program. 2010;2010:101-7.
  • 2. Hodgson DC. Hodgkin lymphoma: the follow-up of long-term survivors. Hematol Oncol Clin North Am. 2008;22(2):23344,vi.
  • 3. Falorio S, Angrilli F, Fioritoni G. Gonadotropin-releasing hormone analog treatment for the prevention of treatment-related ovarian failure and infertility in women of reproductive age with Hodgkin lymphoma. Leuk Lymphoma. 2008;49(6):1087-93.
  • 4. Harel S, Fermé C, Poirot C. Management of fertility in patients treated for Hodgkin lymphoma. Haematologica. 2011;96(11):1692-9.
  • 5. Leader A, Lishner M, Michaeli J, Revel A. Fertility considerations and preservation in haemato-oncology patients undergoing treatment. Br J Haematol. 2011;153(3):291-308.
  • 6 .Faddy MJ, Gosden RG. A model conforming the decline in follicle numbers to the age of menopause in women. Hum Reprod. 1996;11(7):1484-6.
  • 7. Chabbert Buffet N, Djakoure C, Maitre SC, Bouchard P. Regulation of the human menstrual cycle. Front Neuroendocrinol. 1998;19(3):151-86.
  • 8. De Bruin ML, Huisbrink J, Hauptmann M, Kuenen MA, Ouwens GM, van't Veer MB, et al. Treatment-related risk factors for premature menopause following Hodgkin lymphoma. Blood. 2008;111(1):101-8.
  • 9. Wallace WH. Oncofertility and preservation of reproductive capacity in children and young adults. Cancer. 2011;117(10 Suppl):2301-10.
  • 10. Behringer K, Breuer K, Reineke T, May M, Nogova L, Klimm B, et al. Secondary amenorrhea after Hodgkin's lymphoma is influenced by age at treatment, stage of disease, chemotherapy regimen, and the use of oral contraceptives during therapy: a report from the German Hodgkin's Lymphoma Study Group. J Clin Oncol. 2005;23(30):7555-64.
  • 11. Hodgson DC, Pintilie M, Gitterman L, Dewitt B, Buckley CA, Ahmed S, et al. Fertility among female Hodgkin lymphoma survivors attempting pregnancy following ABVD chemotherapy. Hematol Oncol. 2007;25(1):11-5.
  • 12. Loren AW, Chow E, Jacobsohn DA, Gilleece M, Halter J, Joshi S, et al. Pregnancy after hematopoietic cell transplantation: a report from the late effects working committee of the Center for International Blood and Marrow Transplant Research (CIBMTR). Biol Blood Marrow Transplant. 2011;17(2):157-66.
  • 13. Decanter C, Morschhauser F, Pigny P, Lefebvre C, Gallo C, Dewailly D. Anti-Mullerian hormone follow-up in young women treated by chemotherapy for lymphoma: preliminary results. Reprod Biomed Online. 2010;20(2):280-5.
  • 14. Brusamolino E, Baio A, Orlandi E, Arcaini L, Passamonti F, Griva V, et al. Long-term events in adult patients with clinical stage IA-IIA nonbulky Hodgkin's lymphoma treated with four cycles of doxorubicin, bleomycin, vinblastine, and dacarbazine and adjuvant radiotherapy: a single-institution 15-year followup. Clin Cancer Res. 2006;12(21):6487-93.
  • 15. Franchi-Rezgui P, Rousselot P, Espie M, Briere J, Pierre Marolleau J, Gisselbrecht C, et al. Fertility in young women after chemotherapy with alkylating agents for Hodgkin and non-Hodgkin lymphomas. Hematol J. 2003;4(2):116-20.
  • 16. Kiserud CE, Fossa A, Holte H, Fossa SD. Post-treatment parenthood in Hodgkin's lymphoma survivors. Br J Cancer. 2007;96(9):1442-9.
  • 17. Su HI, Chung K, Sammel MD, Gracia CR, DeMichele A. Antral follicle count provides additive information to hormone measures for determining ovarian function in breast cancer survivors. Fertil Steril. 2011;95(5):1857-9.
  • 18. Broekmans FJ, Faddy MJ, Scheffer G, te Velde ER. Antral follicle counts are related to age at natural fertility loss and age at menopause. Menopause. 2004;11(6 Pt 1):607-14. Comment in: Menopause. 2004;11(6 Pt 1):587-8.
  • 19. Rueffer U, Breuer K, Josting A, Lathan B, Sieber M, Manzke O, et al. Male gonadal dysfunction in patients with Hodgkin's disease prior to treatment. Ann Oncol. 2001;12(9):1307-11.
  • 20. Grifo JA, Noyes N. Delivery rate using cryopreserved oocytes is comparable to conventional in vitro fertilization using fresh oocytes: potential fertility preservation for female cancer patients. Fertil Steril. 2010;93(2):391-6.
  • 21. Donnez J, Dolmans MM, Demylle D, Jadoul P, Pirard C, Squifflet J, et al. Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet. 2004;364(9443):1405-10. Comment in: Lancet. 2004;364(9443):1379-80, Lancet. 2004;364(9451):20934, Lancet. 2004;364(9451):2091-2; author reply 2092-3, Lancet. 2004;364(9451):2093. Erratum in: Lancet. 2004;364(9450):2020.
  • 22. Demeestere I, Simon P, Emiliani S, Delbaere A, Englert Y. Fertility preservation: successful transplantation of cryopreserved ovarian tissue in a young patient previously treated for Hodgkin's disease. Oncologist. 2007;12(12):1437-42.
  • 23. Andersen CY, Rosendahl M, Byskov AG, Loft A, Ottosen C, Dueholm M, et al. Two successful pregnancies following autotransplantation of frozen/thawed ovarian tissue. Hum Reprod. 2008;23(10):2266-72.
  • 24. Blumenfeld Z, von Wolff M. GnRH-analogues and oral contraceptives for fertility preservation in women during chemotherapy. Hum Reprod Update. 2008;14(6):543-52. Comment in: Hum Reprod Update. 2008;14(6):541-2.
  • 25. Oktay K, Sonmezer M, Oktem O, Fox K, Emons G, Bang H. Absence of conclusive evidence for the safety and efficacy of gonadotropinreleasing hormone analogue treatment in protecting against chemotherapy-induced gonadal injury. Oncologist. 2007;12(9):1055-66.
  • 26. Blumenfeld Z, Avivi I, Eckman A, Epelbaum R, Rowe JM, Dann EJ. Gonadotropin-releasing hormone agonist decreases chemotherapyinduced gonadotoxicity and premature ovarian failure in young female patients with Hodgkin lymphoma. Fertil Steril. 2008;89(1):166-73. Comment in: Nat Clin Pract Endocrinol Metab. 2008;4(9):488-9, Fertil Steril. 2009;91(1):298-9; author reply 299.
  • 27. Behringer K, Wildt L, Mueller H, Mattle V, Ganitis P, van den Hoonaard B, Ott HW, Hofer S, Pluetschow A, Diehl V, Engert A, Borchmann P, Engert A, Diehl V, Borchmann P, Stein H, Müller RP, Eich HT, Dietlein M, Fuchs M, Behringer K, Böll B, Bredenfeld H, Eichenauer D, Gawlik B, Halbsguth T, Klimm B, v Treskow B, Nisters-Backes H, Tittmann K, Koch B. German Hodgkin Study Group. No protection of the ovarian follicle pool with the use of GnRH-analogues or oral contraceptives in young women treated with escalated BEACOPP for advanced-stage Hodgkin lymphoma. Final results of a phase II trial from the German Hodgkin Study Group. Ann Oncol. 2010;21(10):2052-60. Ann Oncol. 2010;21(10):2052-60.
  • 28. Waxman JH, Ahmed R, Smith D, Wrigley PF, Gregory W, Shalet S, et al. Failure to preserve fertility in patients with Hodgkin's disease. Cancer Chemother Pharmacol. 1987;19(2):159-62.
  • 29. Blumenfeld Z, Avivi I, Linn S, Epelbaum R, Ben-Shahar M, Haim N. Prevention of irreversible chemotherapy-induced ovarian damage in young women with lymphoma by a gonadotrophinreleasing hormone agonist in parallel to chemotherapy. Hum Reprod. 1996;11(8):1620-6.
  • 30. Dann EJ, Epelbaum R, Avivi I, Ben Shahar M, Haim N, Rowe JM, et al. Fertility and ovarian function are preserved in women treated with an intensified regimen of cyclophosphamide, adriamycin, vincristine and prednisone (Mega-CHOP) for non-Hodgkin lymphoma. Hum Reprod. 2005;20(8):2247-9.
  • 31. Huser M, Crha I, Ventruba P, Hudecek R, Zakova J, Smardova L, et al. Prevention of ovarian function damage by a GnRH analogue during chemotherapy in Hodgkin lymphoma patients. Hum Reprod. 2008;23(4):863-8.
  • Corresponding author:

    Massimo Federico
    Department of Oncology, Hematology and respiratory diseases University of Modena and Reggio Emilia Centro Oncologico Modenese
    Via del pozzo
    71 41124 Modena, Italy
    Phone: 39 059 4225515
  • Publication Dates

    • Publication in this collection
      13 Mar 2012
    • Date of issue
      2012

    History

    • Received
      01 Dec 2011
    • Accepted
      04 Dec 2011
    Associação Brasileira de Hematologia e Hemoterapia e Terapia Celular R. Dr. Diogo de Faria, 775 cj 114, 04037-002 São Paulo/SP/Brasil, Tel. (55 11) 2369-7767/2338-6764 - São Paulo - SP - Brazil
    E-mail: secretaria@rbhh.org