Abstract
Tamoxifen (TAM) is a selective estrogen receptor modulator with tissue-specific effects on estrogen signaling used predominantly for treatment and chemoprevention of breast cancers. Recent studies have shown that TAM prevents infertility and decreases follicular loss from common cancer chemotherapy and radiation therapy in preclinical models. Here we review current and novel uses of selective estrogen receptor modulator s and advantages and challenges for translation of TAM for human fertility preservation.
Keywords: cancer, fertility, ovary, SERM, tamoxifen
INTRODUCTION
Recently published preclinical studies demonstrate effective prevention of ovarian toxicity and infertility from cancer chemotherapy drugs and therapeutic radiation using the selective estrogen receptor modulator (SERM) tamoxifen (TAM) as a medical countermeasure [1–3]. This has prompted calls for translation of TAM as an alternative or adjunct to embryonic cryopreservation for fertility preservation in premenopausal women and girls [3]. Here we review the background for this problem, discuss novel uses of SERMs, including as countermeasures to toxicants and drug resistance, and integrate these studies with current clinical indications for TAM.
Current Fertility Preservation in Women and Girls with Cancer
The need for improved fertility in cancer survivors has been recognized repeatedly and reflects improvements in cancer survival. Nearly one million girls and premenopausal women are diagnosed with cancer each year worldwide [4–6]. Standard combination cancer chemotherapy and bone marrow transplantation often cause female infertility and premature menopause [7–9]. The incidence of ovarian failure following cancer treatment varies widely with age and chemotherapy and radiotherapy treatment regimens. Advancing age, dose intensity, cyclophosphamide (Cy), bone marrow transplantation, and abdominal radiation regimens in particular predispose the patients to prolonged amenorrhea following therapy [10–13]. On average, 70% of premenopausal patients receiving alkylating chemotherapy experience prolonged or permanent amenorrhea after treatment [10]. Amenorrhea is less common, but still significant, for other chemotherapy regimens. The impact of cancer chemotherapy on the long-term health of aging women [14] remains largely unaddressed in the literature, but premature menopause is detrimental and associated with heart disease, osteoporosis, stroke, and/or neurological dysfunction [15]. It is important when considering the impact of cancer treatment on fertility to distinguish acute effects of chemotherapy on ovarian function from long-term sequelae such as premature ovarian failure [16].
Embryonic cryopreservation is the current standard of care for fertility preservation in women with cancer wanting children in the future. There are many advantages to this approach. The embryo is removed from exposure to antineoplastics and can be stored for a long period of time for future transfer to the cancer survivor when desired [17, 18]. Over a million children have been born worldwide from cryopreserved embryos. However, this technology requires treatment delay (2–6 wk) and a current partner, is invasive, and fails to protect future ovarian function [19]. Oocyte cryopreservation provides patients without a current partner one method to preserve reproductive potential. Cryopreservation of mature oocytes is no longer considered experimental by the Practice Committees of the American Society for Reproductive Medicine and the Society for Assisted Reproductive Technology [20]. Studies showed no evidence of neonatal risk, chromosomal abnormalities, birth defects, or developmental deficits in babies born from cryopreserved oocytes [21, 22]. Similar to embryo cryopreservation, however, oocyte cryopreservation requires treatment delay and surgical procedures, and does not preserve normal ovarian function. For patients who are prepubertal or require immediate aggressive cancer therapy, ovarian tissue cryopreservation is an option for future fertility [23]. Autografting of frozen-thawed ovarian tissue then provides the only option for restoring ovarian endocrine function and has been shown to induce puberty [24]. However, this technology is still experimental, with only 24 live births born from cryopreserved ovarian tissue [25] since the first case was reported in 2004. Additionally, the infrastructure and high cost associated with cryopreservation limit its widespread use. Because of this, only a small proportion of eligible women seek to avoid reproductive side effects from cancer treatment using this approach.
Alternatives to Cryopreservation for Fertility Preservation in Women with Cancer
Medical countermeasures to ovarian toxicity from cancer treatment have been explored experimentally in females for decades. Broadly speaking, these agents have either been ovarian suppressants or antiapoptotic drugs. Initial attempts used contraceptives to promote ovarian quiescence in an effort to decrease losses of growing ovarian follicles from chemotherapy [26], and there is some evidence of potential benefit in lymphoma patients from this approach [27]. Numerous preclinical and human trials have tested gonadotropin-releasing hormone (GnRH) antagonists for ovarian suppression with mixed results [28–32]. Most recently, Albain and colleagues observed a small but significant increase in posttreatment fertility in estrogen receptor (ER)-negative breast cancer patients supplemented with a GnRH antagonist in the Prevention of Early Menopause Study trial [33]. Importantly, this fertility benefit was accompanied by increased survival, and ER-negative disease is more prevalent in premenopausal women.
Glutathione and sphingosine-1-phosphate are antiapoptotics that have been shown preclinically to decrease follicle loss from antineoplastics and other toxicants [34]. Sphingosine-1-phosphate is the metabolite of ceramide, a sphingolipid second messenger and conserved mediator of germ cell death. These agents do require local ovarian delivery to avoid negative interaction with cancer treatment, but this approach has been demonstrated successfully using a nonhuman primate model [35].
Imatinib, a tyrosine-kinase inhibitor, is commonly used for treatment of chronic myeloid leukemia by inhibiting proliferation and promoting apoptosis in cancer cells. Imatinib targets tyrosine-kinases including BCR-ABL, c-Abl, platelet-derived growth factor (PDGF), receptor as well as c-kit. All of these are associated with cell differentiation, cell division, cell adhesion, and stress response. PDGF and c-kit are expressed in ovarian follicles [36]. Some studies have shown that imatinib can decrease apoptotic follicle loss and infertility resulting from cisplatin and doxorubicin (DOX) [37, 38], but another study contradicts this finding [39]; thus far, imatinib does not appear to affect ovarian toxicity from other agents [38].
Tamoxifen as an Ovarian Protectant
We first observed ovarian protection with TAM incidentally during a preclinical cancer prevention study using 7,12-dimethylbenz [α] anthracene, an experimental carcinogen and ovarian toxicant, to induce ovarian cancer [40]. Although TAM failed to affect ovarian tumorigenesis, TAM-treated rats had greater ovarian follicular reserves than those receiving 7,12-dimethylbenz [α] anthracene alone. In a subsequent study focused on reproductive endpoints, TAM prevented follicle loss and restored fertility in rats receiving Cy regimens that mimic cancer chemotherapy (Fig. 1, adapted from [1]). TAM also improved health and viability of rat cumulus-oocyte-complexes when given with DOX in vitro (Fig. 2, adapted from [1]). Recent work characterized ovarian protection by TAM in vitro. These studies revealed that TAM exerts local antiapoptotic effects in the ovary, and the follicular rescue with TAM was associated with decreased expression of genes related to inflammation [41]. This in vitro study of TAM has established that local ovarian mechanisms of TAM are sufficient for follicular rescue in rodents.
FIG. 1.
A) Hematoxylin and eosin-stained sections of the ovary from rats exposed to vehicle (CONT), cyclophosphamide (Cy, 50 mg/wk), tamoxifen (TAM), or Cy + TAM for 2 wk. Arrowheads = primordial follicles; arrows = primary follicles. Cy decreased follicular reserves compared to CONT, and the addition of TAM inhibited this depletion. TAM alone elevated the number of follicles compared to CONT. Bar = 50 μm. B) Numbers of primordial follicle loss relative to CONT. Average number of follicles per ovary in CONT was 6.1 × 103. Cy at 35 and 50 mg/kg/wk induced follicles loss compared to CONT, and the addition of TAM to both doses of Cy inhibited this depletion. TAM alone did not affect follicle numbers. Different letters indicate significant differences among different treatment groups. Adapted from Ting and Petroff (2010) [1] with permission.
A similar protective effect of TAM on ovarian reserve was also observed by others. Mahran and colleagues demonstrated improved fertility and expanded follicular reserves when using TAM as a countermeasure for ovarian toxicity resulting from therapeutic irradiation in rats [2]. Mechanistically, the most salient feature of this study was the finding that insulin growth factor-1 (IGF-1) receptor was upregulated in the ovary in response to TAM. IGF-1 has been shown previously to play a role in early stage follicle survival in human [42] as well as stimulating granulosa cell function in human and rodents [43, 44]. In addition, IGF-1 is upregulated by ionizing radiation in other tissues [45, 46], but the necessity of this pathway for follicular rescue with TAM remains unknown.
Mechanisms of SERM Action
TAM and other SERMs have a complex and only partially understood mechanism of selective, tissue-specific activation or inhibition of estrogen signaling. The current working model of SERM action and selectivity [47] encompasses 1) competition with endogenous estrogens for ER binding sites, 2) differential affinity and effect of ER isoforms α and β, with α being generally proliferative and β signaling less so, and 3) formation of an altered conformation of the bound receptor in comparison to the natural estrogen ligand leading to altered affinity for estrogen response elements on regulated genes and/or altered recruitment of ER coactivators (for estrogenic SERM actions) or corepressors (for antiestrogenic SERM actions). Of course, it follows that the action of a SERM upon a particular cell type or organ is determined in large part by the complement of ER isoforms, ERE polymorphisms, corepressor, and coactivator plentitude or scarcity and existing concentrations of endogenous estrogens. TAM is largely antiestrogenic in breast tissue while exerting agonistic estrogen-like actions on bone and in the uterus [48]. The impact of TAM on ovarian estrogen signaling is particularly complex because any direct ovarian action is combined with increased gonadotropin stimuli in premenopausal females due to loss of hypothalamic estrogen feedback signaling and because local concentrations of estrogens are remarkably high in the premenopausal ovary [49].
Direct TAM Actions on the Ovary
TAM induces apoptosis in cultured luteal cells in association with elevated bax, bcl-2, and c-myc [50]. Estradiol (E2) and TAM have opposing effects on inhibin expression by rodent granulosa cells in vitro [51], suggesting an antiestrogenic effect of TAM in the ovary, at least for this endpoint. Estradiol-arrested proliferation of granulosa cells not primed with gonadotropin in vitro and TAM reversed this effect [52]. In contrast, both TAM and E2 suppress luteal progesterone production by porcine granulosa cells in vitro [52]. TAM and E2 both suppress proliferation and increase apoptosis of primate ovarian surface epithelium [53]. In vivo, TAM is sometimes used to induce human follicular development in assisted reproductive technology protocols, and such use does not appear to be associated with changes in oocyte quality or maturation [54, 55]. Estradiol has been suggested to act locally in the ovary to increase intercellular communication and gap junction expression in follicular cells and decrease granulosa cell apoptosis [56]. Alteration of these mechanisms of follicular loss and drug exposures are potential mechanisms of follicular rescue by TAM that remain to be tested.
Protective Actions of TAM in Other Tissues
TAM also exerts protective effects in other tissues. Most prominently, TAM decreases tissue damage and loss of function in animal models of spinal cord injury [57, 58] and traumatic or ischemic brain injury [59, 60]. This neuroprotective effect of TAM was associated variously with decreased inflammatory signaling, beneficial redox alterations, increased expression of channel proteins to decrease edema, and diminished NF-KB effects. Interestingly, these benefits from TAM are mimicked by E2 in these tissues [58].
Current Clinical Application of TAM
Therapeutic investigation of TAM has focused on its antagonism of proliferative mammary ER signaling that results in regression of ER-positive breast cancers and reduces the incidence of ER-positive disease by up to 50% when used for prevention of breast cancer in healthy women at increased risk of the disease [61, 62]. Indeed, TAM offers long-term (10-plus yr) benefit against breast cancer [63]. In addition to this established antihormonal benefit, a number of studies have documented, and attempted to exploit in clinical trial, the ability of TAM to reverse multidrug resistance and improve medical outcome. Multidrug resistance to chemotherapy is a major factor in treatment failure and remains a major challenge in cancer therapy [64]. Overexpression of cell membrane pumps, including p-glycoprotein/multidrug resistance-associated proteins, are often found in cancer cells and are responsible for pumping chemotherapy drugs out of the cell, resulting in multidrug resistance [65]. Studies have shown that coadministration of TAM and chemotherapy is associated with downregulation of p-glycoprotein, resulting in potentiation of cytotoxic cancer chemotherapy in a number of cell types [66–68]. This potential effect of tamoxifen was tested in several phase I to III trials with mixed results failing to establish such benefit; nonetheless, results from these trials showed that even high-dose tamoxifen has little effect on the toxicities and pharmacokinetics of standard cancer chemotherapy (Table 1). Interestingly, p-glycoprotein is also expressed in the wall of ovarian capillaries [69] and is responsible for detoxification and steroid transport. Thus, TAM may inhibit chemotherapy transport into the ovary by altering levels of p-glycoproteins.
TABLE 1.
Studies of concurrent TAM and other cancer treatments.a
AEBS, antiestrogen binding site; ADR, adriamycin; DAN, daunorubicin; CAF, cyclophosphamide + doxorubicin + 5-fluorouracil; CDB, cisplatin + dacarbazine + carmustine; CDDP, cisplatin; CEF, cyclophosphamide + epirubicin + 5-fluorouracil; CFP, cyclophosphamide + 5-fluorouracil + prednisone; CHOP, cyclophosphamide + doxorubicin + vincristine + prednisone; CHOPE, cyclophosphamide + doxorubicin + vincristine + prednisone + etoposide; CIS, cisplatin; CMF, cyclophosphamide + methotrexate + 5-fluorouracil; CMFP, CMF + prednisone; CMFV, CMF + vincristine; CTX, cytoxan (cyclophosphamide); EPN, epirubicin; ER, estrogen receptor; 5FU, 5-fluorouracil; IFNβ, interferon β; ISG, IFN-stimulated gene expression; L-PAM, L-phenylalanine mustard; MFL, methotrexate + 5-fluorouracil + leucovorin; MTX, methotrexate; N/A, not applicable; ND, not discussed; PGP, p-glycoprotein; PK, pharmacokinetic, Rad, radiotherapy; TAM, tamoxifen or 4-hydroxytamoxifen (in vitro); TES, tesmilifene; TOR, toremifene; Vin, vinblastine; +, increased; = , no effect; −, decreased.
The Safety of Concurrent Use of TAM with Chemotherapy for Fertility Preservation
Nearly all current use of TAM is for the treatment and prevention of ER-positive (luminal A/B) breast cancer. Treatment protocols can include TAM monotherapy or, more commonly, cytotoxic chemotherapy combinations, such as Cy, methotrexate, and 5-fluorouracil, followed by TAM. Secondary prevention with TAM to prevent recurrence of cancer cells in women who have already undergone treatment for breast cancer after treatment for up to 10 yr is now standard care. More than 12 human trials have examined the impact of concurrently administered TAM and chemotherapy on breast cancer treatment outcomes, including five phase III studies (Table 1). Three of these found no significant impact of concurrent TAM on cancer treatment [70–72]. Mouridsen and colleagues found improved treatment outcomes with concurrent TAM [73]. In the relevant National Surgical Adjuvant Breast and Bowel Project trial, concurrent use of TAM with melphalan and 5-fluorouracil had little impact on most patients. However, worse outcomes were found in the subset of patients with ER-negative/progesterone-negative disease [74]. Two smaller trials comparing sequential versus concurrent TAM and radiation therapy for breast cancer found no difference in clinical outcome between groups [75, 76].
The concurrent use of TAM with combination chemotherapy in breast cancer patients is probably the most demanding (and the most common) clinical scenario for fertility preservation with TAM. The theoretical concern here is that cytostatic actions of TAM on breast tissue will render at least ER-positive breast cancers less sensitive to cytotoxic chemotherapy. As described above, clinical trials do not appear to bear this out and recent trials continue to explore concurrent TAM and chemotherapy as breast cancer therapy [70]. One exception may be hormone receptor-negative breast cancer. The definitive National Surgical Adjuvant Breast and Bowel Project trial did find worse outcomes when TAM was given concurrently with chemotherapy in premenopausal hormone receptor-negative disease [77]. Osborne and colleagues made similar observations in vitro [78]. Interpretation of any fertility data from such trials is further confused by the fact that the majority of patients in these trials were postmenopausal, reflecting the typical patient population for the breast cancer but not the population seeking fertility preservation. Additionally, patients routinely continue to receive TAM for 5 (and now 10) yr after treatment [79], resulting in reversible iatrogenic amenorrhea.
In addition to breast cancer, tamoxifen was shown to improve cancer therapy in vitro in lymphoma and leukemia cell lines by downregulating p-glycoprotein [80]. This led to several human clinical trials in lymphoma and leukemia patients culminating in a phase III trial examining the effect of Cy, hydroxydaunorubicin, oncovin, and prednisone alone or in combination with high-dose TAM [81–84]. These trials found similar outcomes regardless of TAM addition.
Other SERM Candidates for Ovarian Protection During Cancer Treatment
To our knowledge, TAM is the only SERM that has been tested and shown to protect the ovary from cancer treatments. Raloxifene, another SERM, has also been used for ovarian stimulation protocols in assisted reproductive technology and offers similar effects in promoting follicular growth as TAM [85]. In addition, an alternative SERM, tesmilifene, has been tested extensively as a potentiator of chemotherapy action in a number of resistant cancer types [86–88]. In early clinical trials, tesmilifene was shown to increase cytotoxic effects of anthrocyclines (i.e., DOX) [89]. Similar to TAM, tesmilifene enhances cytotoxicity of chemotherapy drugs by selectively targeting p-glycoprotein [90]. Raloxifene and tesmilifene seem to exert TAM-like actions in the ovary and during cancer treatment; however, their potential as an ovarian protectant against chemotherapy has yet to be tested.
SUMMARY
In retrospect, it is not surprising that TAM has selective effects on the ovary. These effects appear to be directly protective to the smallest classes of ovarian follicles—the majority of the follicular reserve. Estrogen receptors, and particularly ERβ, are plentiful in the ovary [91, 92], and protective actions of TAM have been observed for a number of other tissues. The differential expression of ERs, in addition to differences in ER cofactors and local concentrations of other ER ligands, may underlie the apparent ability of TAM to protect ovarian tissue while permitting cancer treatment. While a number of human trials have already tested treatment and survival endpoints when TAM is combined with other antineoplastics or radiation treatment—predominantly with little effect—caution is needed because these populations were mostly postmenopausal and at least one trial found a negative impact of TAM on outcomes for hormone receptor-negative disease. The potential use of TAM for fertility preservation in other tumor types appears more immediately feasible.
TAM is a drug with over 20 million patient-years of use in female cancer patients [93], giving strong safety data in comparison to other medical countermeasures used for fertility preservation. It has been shown preclinically to protect the ovary from a number of follicular toxicants, including cancer treatment drugs cyclophosphamide and doxorubicin as well as therapeutic radiation. Nevertheless, a number of important questions remain unanswered: 1) the mechanism of TAM action during fertility preservation and how this compares with tumoricidal actions in humans have not been established, 2) the necessity of concurrent administration of TAM (vs. pre- or posttreatment) for ovarian protection has not been tested, 3) the effect of other SERMs and aromatase inhibitors are unknown, and 4) the safety of concurrent TAM and chemotherapy administration in breast cancer patients, particularly those with ER-negative disease, remains a question. However, TAM does offer the promise of a convenient, inexpensive, and widely used cancer treatment that can be translated for fertility preservation in some clinical settings, particularly for cancers where TAM may be a potentiator of cancer treatment.
FIG. 2.
A) Mature rat oocytes exposed to vehicle (CONT), 4-hydroxytamoxifen (4HT), doxorubicin (DOX), or DOX+4HT for 26 h. Healthy oocytes surrounded by zona pellucida were observed after CONT and 4HT treatment. DOX induces fragmentation in cultured oocytes, an effect that is antagonized by the addition of 4HT. Original magnification ×400. B) Percentage of oocytes undergoing fragmentation. Oocytes incubated with DOX showed increased fragmentation rate compared with CONT. This elevated fragmentation rate was inhibited by the addition of 4HT. The total number of oocytes cultured in each treatment group is indicated in parentheses inside the respective bar. The asterisk (*) indicates a significant difference in comparison to CONT. Adapted from Ting and Petroff (2010) [1] with permission.
ACKNOWLEDGMENT
The authors would like to thank Dr. John Risinger for critical reading of this manuscript and the Journal of Assisted Reproduction and Genetics for use of figures.
Footnotes
Support was provided by the NIH Center for Biomedical Research Excellence at the University of Kansas Medical Center.
REFERENCES
- Ting AY, Petroff BK. Tamoxifen decreases ovarian follicular loss from experimental toxicant DMBA and chemotherapy agents cyclophosphamide and doxorubicin in the rat. J Assist Reprod Genet. 2010;27:591–597. doi: 10.1007/s10815-010-9463-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahran YF, El-Demerdash E, Nada AS, Ali AA, Abdel-Naim AB. Insights into the protective mechanisms of tamoxifen in radiotherapy-induced ovarian follicular loss: impact on insulin-like growth factor 1. Endocrinology. 2013;154:3888–3899. doi: 10.1210/en.2013-1214. [DOI] [PubMed] [Google Scholar]
- Gleicher N, Kushnir VA, Barad DH. Therapeutic interventions into early stages of follicle maturation: a new treatment paradigm after over 50 years of modern infertility therapy. Endocrinology. 2013;154:3498–3501. doi: 10.1210/en.2013-1679. [DOI] [PubMed] [Google Scholar]
- Falcone T, Attaran M, Bedaiwy MA, Goldberg JM. Ovarian function preservation in the cancer patient. Fertil Steril. 2004;81:243–257. doi: 10.1016/j.fertnstert.2003.06.031. [DOI] [PubMed] [Google Scholar]
- American Cancer Society. Global Cancer Facts & Figures, 2nd ed. Atlanta, GA: American Cancer Society; 2011. [Google Scholar]
- National Cancer Institute Surveilance Epidemiology, and End Results Program; 2011. www.seer.cancer.gov. Accessed 12 December 2014. [Google Scholar]
- Warne GL, Fairley KF, Hobbs JB, Martin FI. Cyclophosphamide-induced ovarian failure. N Engl J Med. 1973;289:1159–1162. doi: 10.1056/NEJM197311292892202. [DOI] [PubMed] [Google Scholar]
- Kim SS. Fertility preservation in female cancer patients: current developments and future directions. Fertil Steril. 2006;85:1–11. doi: 10.1016/j.fertnstert.2005.04.071. [DOI] [PubMed] [Google Scholar]
- Schmidt KT, Larsen EC, Andersen CY, Andersen AN. Risk of ovarian failure and fertility preserving methods in girls and adolescents with a malignant disease. BJOG. 2010;117:163–174. doi: 10.1111/j.1471-0528.2009.02408.x. [DOI] [PubMed] [Google Scholar]
- Bines J, Oleske DM, Cobleigh MA. Ovarian function in premenopausal women treated with adjuvant chemotherapy for breast cancer. J Clin Oncol. 1996;14:1718–1729. doi: 10.1200/JCO.1996.14.5.1718. [DOI] [PubMed] [Google Scholar]
- Meirow D. Reproduction post-chemotherapy in young cancer patients. Mol Cell Endocrinol. 2000;169:123–131. doi: 10.1016/s0303-7207(00)00365-8. [DOI] [PubMed] [Google Scholar]
- Barton SE, Najita JS, Ginsburg ES, Leisenring WM, Stovall M, Weathers RE, Sklar CA, Robison LL, Diller L. Infertility, infertility treatment, and achievement of pregnancy in female survivors of childhood cancer: a report from the Childhood Cancer Survivor Study cohort. Lancet Oncol. 2013;14:873–881. doi: 10.1016/S1470-2045(13)70251-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blumenfeld Z, Eckman A. Preservation of fertility and ovarian function and minimalization of chemotherapy associated gonadotoxicity and premature ovarian failure: the role of inhibin-A and -B as markers. Mol Cell Endocrinol. 2002;187:93–105. doi: 10.1016/s0303-7207(01)00712-2. [DOI] [PubMed] [Google Scholar]
- Shuster LT, Rhodes DJ, Gostout BS, Grossardt BR, Rocca WA. Premature menopause or early menopause: long-term health consequences. Maturitas. 2010;65:161–166. doi: 10.1016/j.maturitas.2009.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oktay K, Sonmezer M. Chemotherapy and amenorrhea: risks and treatment options. Curr Opin Obstet Gynecol. 2008;20:408–415. doi: 10.1097/GCO.0b013e328307ebad. [DOI] [PubMed] [Google Scholar]
- Morgan S, Anderson RA, Gourley C, Wallace WH, Spears N. How do chemotherapeutic agents damage the ovary? Hum Reprod Update. 2012;18:525–535. doi: 10.1093/humupd/dms022. [DOI] [PubMed] [Google Scholar]
- Check JH, Summers-Chase D, Yuan W, Swenson K, Horwath D. Length of time of embryo storage does not negatively influence pregnancy rates after thawing and transfer. Clin Exp Obstet Gynecol. 2010;37:185–186. [PubMed] [Google Scholar]
- Riggs R, Mayer J, Dowling-Lacey D, Chi TF, Jones E, Oehninger S. Does storage time influence postthaw survival and pregnancy outcome? An analysis of 11,768 cryopreserved human embryos. Fertil Steril. 2010;93:109–115. doi: 10.1016/j.fertnstert.2008.09.084. [DOI] [PubMed] [Google Scholar]
- Harel S, Ferme C, Poirot C. Management of fertility in patients treated for Hodgkin's lymphoma. Haematologica. 2011;96:1692–1699. doi: 10.3324/haematol.2011.045856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- The Practice Committee of the American Society for Reproductive Medicine and the Society for Assisted Reproductive Technology. Mature oocyte cryopreservation: a guideline. Fertil Steril. 2013;99:37–43. doi: 10.1016/j.fertnstert.2012.09.028. [DOI] [PubMed] [Google Scholar]
- Cobo A, Rubio C, Gerli S, Ruiz A, Pellicer A, Remohi J. Use of fluorescence in situ hybridization to assess the chromosomal status of embryos obtained from cryopreserved oocytes. Fertil Steril. 2001;75:354–360. doi: 10.1016/s0015-0282(00)01725-8. [DOI] [PubMed] [Google Scholar]
- Noyes N, Porcu E, Borini A. Over 900 oocyte cryopreservation babies born with no apparent increase in congenital anomalies. Reprod Biomed Online. 2009;18:769–776. doi: 10.1016/s1472-6483(10)60025-9. [DOI] [PubMed] [Google Scholar]
- Jeruss JS, Woodruff TK. Preservation of fertility in patients with cancer. N Engl J Med. 2009;360:902–911. doi: 10.1056/NEJMra0801454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poirot C, Abirached F, Prades M, Coussieu C, Bernaudin F, Piver P. Induction of puberty by autograft of cryopreserved ovarian tissue. Lancet. 2012;379:588. doi: 10.1016/S0140-6736(11)61781-9. [DOI] [PubMed] [Google Scholar]
- Donnez J, Dolmans MM, Pellicer A, Diaz-Garcia C, Sanchez Serrano M, Schmidt KT, Ernst E, Luyckx V, Andersen CY. Restoration of ovarian activity and pregnancy after transplantation of cryopreserved ovarian tissue: a review of 60 cases of reimplantation. Fertil Steril. 2013;99:1503–1513. doi: 10.1016/j.fertnstert.2013.03.030. [DOI] [PubMed] [Google Scholar]
- Chapman RM, Sutcliffe SB. Protection of ovarian function by oral contraceptives in women receiving chemotherapy for Hodgkin's disease. Blood. 1981;58:849–851. [PubMed] [Google Scholar]
- Behringer K, Breuer K, Reineke T, May M, Nogova L, Klimm B, Schmitz T, Wildt L, Diehl V, Engert A. 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;23:7555–7564. doi: 10.1200/JCO.2005.08.138. [DOI] [PubMed] [Google Scholar]
- Meistrich ML, Shetty G. Hormonal suppression for fertility preservation in males and females. Reproduction. 2008;136:691–701. doi: 10.1530/REP-08-0096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karimi-Zarchi M, Forat-Yazdi M, Vafaeenasab MR, Nakhaie-Moghadam M, Miratashi-Yazdi A, Teimoori S, Dehghani-Tafti A. Evaluation of the effect of GnRH agonist on menstrual reverse in breast cancer cases treated with cyclophosphamide. Eur J Gynaecol Oncol. 2014;35:59–61. [PubMed] [Google Scholar]
- Vitek WS, Shayne M, Hoeger K, Han Y, Messing S, Fung C. Gonadotropin-releasing hormone agonists for the preservation of ovarian function among women with breast cancer who did not use tamoxifen after chemotherapy: a systematic review and meta-analysis. Fertil Steril. 2014;102:808–815. doi: 10.1016/j.fertnstert.2014.06.003. [DOI] [PubMed] [Google Scholar]
- Del Mastro L, Ceppi M, Poggio F, Bighin C, Peccatori F, Demeestere I, Levaggi A, Giraudi S, Lambertini M, D'Alonzo A, Canavese G, Pronzato P, et al. Gonadotropin-releasing hormone analogues for the prevention of chemotherapy-induced premature ovarian failure in cancer women: systematic review and meta-analysis of randomized trials. Cancer Treat Rev. 2014;40:675–683. doi: 10.1016/j.ctrv.2013.12.001. [DOI] [PubMed] [Google Scholar]
- Partridge AH. Ovarian suppression for prevention of premature menopause and infertility: empty promise or effective therapy? J Clin Oncol. 2012;30:479–481. doi: 10.1200/JCO.2011.37.9883. [DOI] [PubMed] [Google Scholar]
- Moore HCF, Unger JM, Phillips K, Boyle FM, Hitre E, Porter DJ, Francis PA, Minasian LM, Gelber RD, Goldstein LJ, Gomez HL, Vallejos C, et al. Phase III trial (Prevention of Early Menopause Study [POEMS]-SWOG S0230) of LHRH analog during chemotherapy (CT) to reduce ovarian failure in early-stage, hormone receptor-negative breast cancer: An international Intergroup trial of SWOG, IBCSG, ECOG, and CALGB (Alliance) J Clin Oncol 2014. 32 (suppl) abstr LBA505. [Google Scholar]
- Morita Y, Perez GI, Paris F, Miranda SR, Ehleiter D, Haimovitz-Friedman A, Fuks Z, Xie Z, Reed JC, Schuchman EH, Kolesnick RN, Tilly JL. Oocyte apoptosis is suppressed by disruption of the acid sphingomyelinase gene or by sphingosine-1-phosphate therapy. Nat Med. 2000;6:1109–1114. doi: 10.1038/80442. [DOI] [PubMed] [Google Scholar]
- Zelinski MB, Murphy MK, Lawson MS, Jurisicova A, Pau KY, Toscano NP, Jacob DS, Fanton JK, Casper RF, Dertinger SD, Tilly JL. In vivo delivery of FTY720 prevents radiation-induced ovarian failure and infertility in adult female nonhuman primates. Fertil Steril. 2011;95:1440–1445. doi: 10.1016/j.fertnstert.2011.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nilsson EE, Detzel C, Skinner MK. Platelet-derived growth factor modulates the primordial to primary follicle transition. Reproduction. 2006;131:1007–1015. doi: 10.1530/rep.1.00978. [DOI] [PubMed] [Google Scholar]
- Morgan S, Lopes F, Gourley C, Anderson RA, Spears N. Cisplatin and doxorubicin induce distinct mechanisms of ovarian follicle loss; imatinib provides selective protection only against cisplatin. PLoS One. 2013;8:e70117. doi: 10.1371/journal.pone.0070117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonfloni S, Di Tella L, Caldarola S, Cannata SM, Klinger FG, Di Bartolomeo C, Mattei M, Candi E, De Felici M, Melino G, Cesareni G. Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapy-induced death. Nat Med. 2009;15:1179–1185. doi: 10.1038/nm.2033. [DOI] [PubMed] [Google Scholar]
- Kerr JB, Hutt KJ, Cook M, Speed TP, Strasser A, Findlay JK, Scott CL. Cisplatin-induced primordial follicle oocyte killing and loss of fertility are not prevented by imatinib Nat Med 2012. 18 1170 1172 (author reply 1172–1174). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ting AY, Kimler BF, Fabian CJ, Petroff BK. Tamoxifen prevents premalignant changes of breast, but not ovarian, cancer in rats at high risk for both diseases. Cancer Prev Res (Phila) 2008;1:546–553. doi: 10.1158/1940-6207.CAPR-08-0015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piasecka-Srader J, Blanco FF, Delman DH, Dixon DA, Geiser JL, Ciereszko RE, Petroff BK. Tamoxifen prevents apoptosis and follicle loss from cyclophosphamide in cultured rat ovaries. Biol Reprod. 2015;92:132. doi: 10.1095/biolreprod.114.126136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Louhio H, Hovatta O, Sjoberg J, Tuuri T. The effects of insulin, and insulin-like growth factors I and II on human ovarian follicles in long-term culture. Mol Hum Reprod. 2000;6:694–698. doi: 10.1093/molehr/6.8.694. [DOI] [PubMed] [Google Scholar]
- Baumgarten SC, Convissar SM, Fierro MA, Winston NJ, Scoccia B, Stocco C. IGF1R signaling is necessary for FSH-induced activation of AKT and differentiation of human Cumulus granulosa cells. J Clin Endocrinol Metab. 2014;99:2995–3004. doi: 10.1210/jc.2014-1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou P, Baumgarten SC, Wu Y, Bennett J, Winston N, Hirshfeld-Cytron J, Stocco C. IGF-I signaling is essential for FSH stimulation of AKT and steroidogenic genes in granulosa cells. Mol Endocrinol. 2013;27:511–523. doi: 10.1210/me.2012-1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cosaceanu D, Budiu RA, Carapancea M, Castro J, Lewensohn R, Dricu A. Ionizing radiation activates IGF-1R triggering a cytoprotective signaling by interfering with Ku-DNA binding and by modulating Ku86 expression via a p38 kinase-dependent mechanism. Oncogene. 2007;26:2423–2434. doi: 10.1038/sj.onc.1210037. [DOI] [PubMed] [Google Scholar]
- Floratou K, Giannopoulou E, Antonacopoulou A, Karakantza M, Adonakis G, Kardamakis D, Matsouka P. Oxidative stress due to radiation in CD34(+) hematopoietic progenitor cells: protection by IGF-1. J Radiat Res. 2012;53:672–685. doi: 10.1093/jrr/rrs019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jordan VC, O'Malley BW. Selective estrogen-receptor modulators and antihormonal resistance in breast cancer. J Clin Oncol. 2007;25:5815–5824. doi: 10.1200/JCO.2007.11.3886. [DOI] [PubMed] [Google Scholar]
- Shang Y, Brown M. Molecular determinants for the tissue specificity of SERMs. Science. 2002;295:2465–2468. doi: 10.1126/science.1068537. [DOI] [PubMed] [Google Scholar]
- Thuesen LL, Andersen AN, Loft A, Smitz J. Intrafollicular endocrine milieu after addition of hCG to recombinant FSH during controlled ovarian stimulation for in vitro fertilization. J Clin Endocrinol Metab. 2014;99:517–526. doi: 10.1210/jc.2013-1528. [DOI] [PubMed] [Google Scholar]
- Leng Y, Gu ZP, Cao L. Apoptosis induced by droloxifene and c-myc, bax and bcl-2 mRNA expression in cultured luteal cells of rats. Eur J Pharmacol. 2000;409:123–131. doi: 10.1016/s0014-2999(00)00826-8. [DOI] [PubMed] [Google Scholar]
- Tate S, Suganuma N, Furuhashi M, Ando T, Asada Y, Kondo I, Kikkawa F, Tomoda Y. Direct effects of estradiol and tamoxifen on gene expressions of inhibit alpha- and beta A-subunits in rat granulosa cells in vitro. Endocr J. 1996;43:621–628. doi: 10.1507/endocrj.43.621. [DOI] [PubMed] [Google Scholar]
- Luciano AM, Peluso JJ. Effect of in vivo gonadotropin treatment on the ability of progesterone, estrogen, and cyclic adenosine 5′-monophosphate to inhibit insulin-dependent granulosa cell mitosis in vitro. Biol Reprod. 1995;53:664–669. doi: 10.1095/biolreprod53.3.664. [DOI] [PubMed] [Google Scholar]
- Wright JW, Stouffer RL, Rodland KD. High-dose estrogen and clinical selective estrogen receptor modulators induce growth arrest, p21, and p53 in primate ovarian surface epithelial cells. J Clin Endocrinol Metab. 2005;90:3688–3695. doi: 10.1210/jc.2004-2456. [DOI] [PubMed] [Google Scholar]
- Fisk NM, Templeton AA, Papadopoulos GC, Matlin SA, Wu ZY. Lack of effect of high-dose antioestrogen on the maturation and in-vitro fertilization of human oocytes. Hum Reprod. 1989;4:584–587. doi: 10.1093/oxfordjournals.humrep.a136947. [DOI] [PubMed] [Google Scholar]
- Meirow D, Raanani H, Maman E, Paluch-Shimon S, Shapira M, Cohen Y, Kuchuk I, Hourvitz A, Levron J, Mozer-Mendel M, Brengauz M, Biderman H, et al. Tamoxifen co-administration during controlled ovarian hyperstimulation for in vitro fertilization in breast cancer patients increases the safety of fertility-preservation treatment strategies. Fertil Steril. 2014;102:488–495. doi: 10.1016/j.fertnstert.2014.05.017. [DOI] [PubMed] [Google Scholar]
- Rosenfeld CS, Wagner JS, Roberts RM, Lubahn DB. Intraovarian actions of oestrogen. Reproduction. 2001;122:215–226. doi: 10.1530/rep.0.1220215. [DOI] [PubMed] [Google Scholar]
- Tian DS, Liu JL, Xie MJ, Zhan Y, Qu WS, Yu ZY, Tang ZP, Pan DJ, Wang W. Tamoxifen attenuates inflammatory-mediated damage and improves functional outcome after spinal cord injury in rats. J Neurochem. 2009;109:1658–1667. doi: 10.1111/j.1471-4159.2009.06077.x. [DOI] [PubMed] [Google Scholar]
- Guptarak J, Wiktorowicz JE, Sadygov RG, Zivadinovic D, Paulucci-Holthauzen AA, Vergara L, Nesic O. The cancer drug tamoxifen: a potential therapeutic treatment for spinal cord injury. J Neurotrauma. 2014;31:268–283. doi: 10.1089/neu.2013.3108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wakade C, Khan MM, De Sevilla LM, Zhang QG, Mahesh VB, Brann DW. Tamoxifen neuroprotection in cerebral ischemia involves attenuation of kinase activation and superoxide production and potentiation of mitochondrial superoxide dismutase. Endocrinology. 2008;149:367–379. doi: 10.1210/en.2007-0899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun X, Ji C, Hu T, Wang Z, Chen G. Tamoxifen as an effective neuroprotectant against early brain injury and learning deficits induced by subarachnoid hemorrhage: possible involvement of inflammatory signaling. J Neuroinflamm. 2013;10:157. doi: 10.1186/1742-2094-10-157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jordan VC. Tamoxifen (ICI46,474) as a targeted therapy to treat and prevent breast cancer. Br J Pharmacol. 2006;147((Suppl 1)):S269–S276. doi: 10.1038/sj.bjp.0706399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Errico A. Breast cancer: tamoxifen-offering a long-term prevention option. Nat Rev Clin Oncol. 2015;12:66. doi: 10.1038/nrclinonc.2014.236. [DOI] [PubMed] [Google Scholar]
- Sestak I, Cuzick J. Update on breast cancer risk prediction and prevention. Curr Opin Obstet Gynecol. 2015;27:92–97. doi: 10.1097/GCO.0000000000000153. [DOI] [PubMed] [Google Scholar]
- Persidis A. Cancer multidrug resistance. Nat Biotechnol. 1999;17:94–95. doi: 10.1038/5289. [DOI] [PubMed] [Google Scholar]
- Pastan I, Gottesman MM. Multidrug resistance. Annu Rev Med. 1991;42:277–286. doi: 10.1146/annurev.me.42.020191.001425. [DOI] [PubMed] [Google Scholar]
- Callaghan R, Higgins CF. Interaction of tamoxifen with the multidrug resistance P-glycoprotein. Br J Cancer. 1995;71:294–299. doi: 10.1038/bjc.1995.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu ZH, Ma YL, He YP, Zhang P, Zhou YK, Qin H. Tamoxifen reverses the multi-drug-resistance of an established human cholangiocarcinoma cell line in combined chemotherapeutics. Mol Biol Rep. 2011;38:1769–1775. doi: 10.1007/s11033-010-0291-z. [DOI] [PubMed] [Google Scholar]
- Chapman JV, Gouaze-Andersson V, Karimi R, Messner MC, Cabot MC. P-glycoprotein antagonists confer synergistic sensitivity to short-chain ceramide in human multidrug-resistant cancer cells. Exp Cell Res. 2011;317:1736–1745. doi: 10.1016/j.yexcr.2011.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stewart PA, Beliveau R, Rogers KA. Cellular localization of P-glycoprotein in brain versus gonadal capillaries. J Histochem Cytochem. 1996;44:679–685. doi: 10.1177/44.7.8675989. [DOI] [PubMed] [Google Scholar]
- Bedognetti D, Sertoli MR, Pronzato P, Del Mastro L, Venturini M, Taveggia P, Zanardi E, Siffredi G, Pastorino S, Queirolo P, Gardin G, Wang E, et al. Concurrent vs sequential adjuvant chemotherapy and hormone therapy in breast cancer: a multicenter randomized phase III trial. J Natl Cancer Inst. 2011;103:1529–1539. doi: 10.1093/jnci/djr351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pico C, Martin M, Jara C, Barnadas A, Pelegri A, Balil A, Camps C, Frau A, Rodriguez-Lescure A, Lopez-Vega JM, De La Haba J, Tres A, et al. Epirubicin-cyclophosphamide adjuvant chemotherapy plus tamoxifen administered concurrently versus sequentially: randomized phase III trial in postmenopausal node-positive breast cancer patients. A GEICAM 9401 study. Ann Oncol. 2004;15:79–87. doi: 10.1093/annonc/mdh016. [DOI] [PubMed] [Google Scholar]
- Albain KS, Barlow WE, Ravdin PM, Farrar WB, Burton GV, Ketchel SJ, Cobau CD, Levine EG, Ingle JN, Pritchard KI, Lichter AS, Schneider DJ, et al. Adjuvant chemotherapy and timing of tamoxifen in postmenopausal patients with endocrine-responsive, node-positive breast cancer: a phase 3, open-label, randomised controlled trial. Lancet. 2009;374:2055–2063. doi: 10.1016/S0140-6736(09)61523-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mouridsen HT, Rose C, Engelsmann E, Sylvester R, Rotmensz N. Combined cytotoxic and endocrine therapy in postmenopausal patients with advanced breast cancer. A randomized EORTC study of CMF vs CMF + tamoxifen. J Steroid Biochem. 1985;23:1141–1146. doi: 10.1016/0022-4731(85)90033-0. [DOI] [PubMed] [Google Scholar]
- Taylor SG IV, Kalish LA, Olson JE, Cummings F, Bennett JM, Falkson G, Tormey DC, Carbone PP. Adjuvant CMFP versus CMFP plus tamoxifen versus observation alone in postmenopausal, node-positive breast cancer patients: three-year results of an Eastern Cooperative Oncology Group study. J Clin Oncol. 1985;3:144–154. doi: 10.1200/JCO.1985.3.2.144. [DOI] [PubMed] [Google Scholar]
- Harris EE, Christensen VJ, Hwang WT, Fox K, Solin LJ. Impact of concurrent versus sequential tamoxifen with radiation therapy in early-stage breast cancer patients undergoing breast conservation treatment. J Clin Oncol. 2005;23:11–16. doi: 10.1200/JCO.2005.09.056. [DOI] [PubMed] [Google Scholar]
- Pierce LJ, Hutchins LF, Green SR, Lew DL, Gralow JR, Livingston RB, Osborne CK, Albain KS. Sequencing of tamoxifen and radiotherapy after breast-conserving surgery in early-stage breast cancer. J Clin Oncol. 2005;23:24–29. doi: 10.1200/JCO.2005.01.198. [DOI] [PubMed] [Google Scholar]
- Fisher B, Redmond C, Brown A, Fisher ER, Wolmark N, Bowman D, Plotkin D, Wolter J, Bornstein R, Legault-Poisson S, Saffer EA. Adjuvant chemotherapy with and without tamoxifen in the treatment of primary breast cancer: 5-year results from the National Surgical Adjuvant Breast and Bowel Project Trial. J Clin Oncol. 1986;4:459–471. doi: 10.1200/JCO.1986.4.4.459. [DOI] [PubMed] [Google Scholar]
- Osborne CK, Kitten L, Arteaga CL. Antagonism of chemotherapy-induced cytotoxicity for human breast cancer cells by antiestrogens. J Clin Oncol. 1989;7:710–717. doi: 10.1200/JCO.1989.7.6.710. [DOI] [PubMed] [Google Scholar]
- Davies C, Pan H, Godwin J, Gray R, Arriagada R, Raina V, Abraham M, Medeiros Alencar VH, Badran A, Bonfill X, Bradbury J, Clarke M, et al. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet. 2013;381:805–816. doi: 10.1016/S0140-6736(12)61963-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berman E, Adams M, Duigou-Osterndorf R, Godfrey L, Clarkson B, Andreeff M. Effect of tamoxifen on cell lines displaying the multidrug-resistant phenotype. Blood. 1991;77:818–825. [PubMed] [Google Scholar]
- Berman E, McBride M, Lin S, Menedez-Botet C, Tong W. Phase I trial of high-dose tamoxifen as a modulator of drug resistance in combination with daunorubicin in patients with relapsed or refractory acute leukemia. Leukemia. 1995;9:1631–1637. [PubMed] [Google Scholar]
- El-Yazigi A, Berry J, Ezzat A, Wahab FA. Effect of tamoxifen on the pharmacokinetics of doxorubicin in patients with non-Hodgkin's lymphoma. Ther Drug Monit. 1997;19:632–636. doi: 10.1097/00007691-199712000-00005. [DOI] [PubMed] [Google Scholar]
- Ezzat AA, Ibrahim EM, Stuart RK, Ajarim D, Bazarbashi S, El-Foudeh MO, Rahal M, Al-Sayed A, Berry J. Adding high-dose tamoxifen to CHOP does not influence response or survival in aggressive non-Hodgkin's lymphoma: an interim analysis of a randomized phase III trial. Med Oncol. 2000;17:39–46. doi: 10.1007/BF02826215. [DOI] [PubMed] [Google Scholar]
- Al-Gailani F, Marder RJ, Radosevich JA, Hauck WW, Molteni A, Rosen ST. Phase II trial of tamoxifen in chronic lymphocytic leukemia. Cancer Treat Rep. 1986;70:1343–1344. [PubMed] [Google Scholar]
- Premkumar A, Venzon DJ, Avila N, Johnson DV, Remaley AT, Forman MR, Eng-Wong J, Zujewski J, Stratton P. Gynecologic and hormonal effects of raloxifene in premenopausal women. Fertil Steril. 2007;88:1637–1644. doi: 10.1016/j.fertnstert.2007.01.095. [DOI] [PubMed] [Google Scholar]
- Brandes LJ. N,N-diethyl-2-[4-(phenylmethyl) phenoxy] ethanamine (DPPE; tesmilifene), a chemopotentiating agent with hormetic effects on DNA synthesis in vitro, may improve survival in patients with metastatic breast cancer. Hum Exp Toxicol. 2008;27:143–147. doi: 10.1177/0960327108090751. [DOI] [PubMed] [Google Scholar]
- Choi YH, Suh JH, Lee JH, Cho IH, Lee MG. Effects of tesmilifene, a substrate of CYP3A and an inhibitor of P-glycoprotein, on the pharmacokinetics of intravenous and oral docetaxel in rats. J Pharm Pharmacol. 2010;62:1084–1088. doi: 10.1111/j.2042-7158.2010.01129.x. [DOI] [PubMed] [Google Scholar]
- Ferguson PJ, Brisson AR, Koropatnick J, Vincent MD. Enhancement of cytotoxicity of natural product drugs against multidrug resistant variant cell lines of human head and neck squamous cell carcinoma and breast carcinoma by tesmilifene. Cancer Lett. 2009;274:279–289. doi: 10.1016/j.canlet.2008.09.021. [DOI] [PubMed] [Google Scholar]
- Reyno L, Seymour L, Tu D, Dent S, Gelmon K, Walley B, Pluzanska A, Gorbunova V, Garin A, Jassem J, Pienkowski T, Dancey J, et al. Phase III study of N,N-diethyl-2-[4-(phenylmethyl) phenoxy]ethanamine (BMS-217380-01) combined with doxorubicin versus doxorubicin alone in metastatic/recurrent breast cancer: National Cancer Institute of Canada Clinical Trials Group Study MA.19. J Clin Oncol. 2004;22:269–276. doi: 10.1200/JCO.2003.04.075. [DOI] [PubMed] [Google Scholar]
- Georges E, Lian J, Laberge R. A tamoxifen derivative, N,N-diethyl-2-[4-(phenylmethyl) phenoxy] ethanamine, selectively targets P-glycoprotein-positive multidrug resistant Chinese hamster cells. Biochem Pharmacol. 2014;90:107–114. doi: 10.1016/j.bcp.2014.04.017. [DOI] [PubMed] [Google Scholar]
- Saunders PT, Millar MR, Williams K, Macpherson S, Harkiss D, Anderson RA, Orr B, Groome NP, Scobie G, Fraser HM. Differential expression of estrogen receptor-alpha and -beta and androgen receptor in the ovaries of marmosets and humans. Biol Reprod. 2000;63:1098–1105. doi: 10.1095/biolreprod63.4.1098. [DOI] [PubMed] [Google Scholar]
- Pelletier G, El-Alfy M. Immunocytochemical localization of estrogen receptors alpha and beta in the human reproductive organs. J Clin Endocrinol Metab. 2000;85:4835–4840. doi: 10.1210/jcem.85.12.7029. [DOI] [PubMed] [Google Scholar]
- Wickerham DL. Association of tamoxifen and uterine sarcoma. J Clin Oncol. 2002;20:2758–2760. doi: 10.1200/JCO.2002.20.11.2758. [DOI] [PubMed] [Google Scholar]
- Boccardo F, Rubagotti A, Rosso R, Santi L. Chemotherapy with or without tamoxifen in postmenopausal patients with late breast cancer. A randomized study. J Steroid Biochem. 1985;23:1123–1127. doi: 10.1016/0022-4731(85)90030-5. [DOI] [PubMed] [Google Scholar]
- Crowe PJ, Jr, Gordon NH, Shenk RR, Soegiarso RW, Hubay CA, Mansour EG, Shuck JM, Pearson OH, Marshall JS, Arafah B, et al. Short-term tamoxifen plus chemotherapy: superior results in node-positive breast cancer Surgery 1990. 108 619 627 (discussion 627–628). [PubMed] [Google Scholar]
- Ingle JN, Everson LK, Wieand HS, Martin JK, Votava HJ, Wold LE, Krook JE, Cullinan SA, Paulsen JK, Twito DI. Randomized trial of observation versus adjuvant therapy with cyclophosphamide, fluorouracil, prednisone with or without tamoxifen following mastectomy in postmenopausal women with node-positive breast cancer. J Clin Oncol. 1988;6:1388–1396. doi: 10.1200/JCO.1988.6.9.1388. [DOI] [PubMed] [Google Scholar]
- Ingle JN, Everson LK, Wieand HS, Cullinan SA, Wold LE, Hagen JB, Martin JK, Krook JE, Fitzgibbons RG, Foley JF. Randomized trial to evaluate the addition of tamoxifen to cyclophosphamide, 5-fluorouracil, prednisone adjuvant therapy in premenopausal women with node-positive breast cancer. Cancer. 1989;63:1257–1264. doi: 10.1002/1097-0142(19890401)63:7<1257::aid-cncr2820630705>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
- CMEA Cooperative Treatment Group. Clinical controlled trial in advanced breast cancer: CMF (cyclophosphamide, methotrexate, 5-fluorouracil) versus CMF + T (cyclophosphamide, methotrexate, 5-fluorouracil, tamoxifen) Neoplasma. 1985;32:381–387. [PubMed] [Google Scholar]
- Krook JE, Ingle JN, Green SJ, Bowman WD, Everson LK, Windschitl HE, Marschke RF, Laurie JA, Cullinan SA, Pfeifle DM. Randomized clinical trial of cyclophosphamide, 5-FU, and prednisone with or without tamoxifen in postmenopausal women with advanced breast cancer. Cancer Treat Rep. 1985;69:355–361. [PubMed] [Google Scholar]
- Lindner DJ, Borden EC. Synergistic antitumor effects of a combination of interferon and tamoxifen on estrogen receptor-positive and receptor-negative human tumor cell lines in vivo and in vitro. J Interferon Cytokine Res. 1997;17:681–693. doi: 10.1089/jir.1997.17.681. [DOI] [PubMed] [Google Scholar]
- Woods KE, Randolph JK, Gewirtz DA. Antagonism between tamoxifen and doxorubicin in the MCF-7 human breast tumor cell line. Biochem Pharmacol. 1994;47:1449–1452. doi: 10.1016/0006-2952(94)90346-8. [DOI] [PubMed] [Google Scholar]
- Hug V, Hortobagyi GN, Drewinko B, Finders M. Tamoxifen-citrate counteracts the antitumor effects of cytotoxic drugs in vitro. J Clin Oncol. 1985;3:1672–1677. doi: 10.1200/JCO.1985.3.12.1672. [DOI] [PubMed] [Google Scholar]
- Benz C, Cadman E, Gwin J, Wu T, Amara J, Eisenfeld A, Dannies P. Tamoxifen and 5-fluorouracil in breast cancer: cytotoxic synergism in vitro. Cancer Res. 1983;43:5298–5303. [PubMed] [Google Scholar]
- Kurebayashi J, Nukatsuka M, Nagase H, Nomura T, Hirono M, Yamamoto Y, Sugimoto Y, Oka T, Sonoo H. Additive antitumor effect of concurrent treatment of 4-hydroxy tamoxifen with 5-fluorouracil but not with doxorubicin in estrogen receptor-positive breast cancer cells. Cancer Chemother Pharmacol. 2007;59:515–525. doi: 10.1007/s00280-006-0293-7. [DOI] [PubMed] [Google Scholar]
- Chatterjee M, Harris AL. Reversal of acquired resistance to adriamycin in CHO cells by tamoxifen and 4-hydroxy tamoxifen: role of drug interaction with alpha 1 acid glycoprotein. Br J Cancer. 1990;62:712–717. doi: 10.1038/bjc.1990.365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trump DL, Smith DC, Ellis PG, Rogers MP, Schold SC, Winer EP, Panella TJ, Jordan VC, Fine RL. High-dose oral tamoxifen, a potential multidrug-resistance-reversal agent: phase I trial in combination with vinblastine. J Natl Cancer Inst. 1992;84:1811–1816. doi: 10.1093/jnci/84.23.1811. [DOI] [PubMed] [Google Scholar]
- Mao Z, Zhou J, Luan J, Sheng W, Shen X, Dong X. Tamoxifen reduces P-gp-mediated multidrug resistance via inhibiting the PI3K/Akt signaling pathway in ER-negative human gastric cancer cells. Biomed Pharmacother. 2014;68:179–183. doi: 10.1016/j.biopha.2013.10.003. [DOI] [PubMed] [Google Scholar]
- Smith DC, Trump DL. A phase I trial of high-dose oral tamoxifen and CHOPE. Cancer Chemother Pharmacol. 1995;36:65–68. doi: 10.1007/BF00685734. [DOI] [PubMed] [Google Scholar]
- Creagan ET, Suman VJ, Dalton RJ, Pitot HC, Long HJ, Veeder MH, Vukov AM, Rowland KM, Krook JE, Michalak JC. Phase III clinical trial of the combination of cisplatin, dacarbazine, and carmustine with or without tamoxifen in patients with advanced malignant melanoma. J Clin Oncol. 1999;17:1884–1890. doi: 10.1200/JCO.1999.17.6.1884. [DOI] [PubMed] [Google Scholar]
- Jones JA, Albright KD, Christen RD, Howell SB, McClay EF. Synergy between tamoxifen and cisplatin in human melanoma cells is dependent on the presence of antiestrogen-binding sites. Cancer Res. 1997;57:2657–2660. [PubMed] [Google Scholar]
- Ouyang ZX, Li XA. Inhibitory effects of tamoxifen and doxorubicin, alone and in combination, on the proliferation of the MG63 human osteosarcoma cell line. Oncol Lett. 2013;6:970–976. doi: 10.3892/ol.2013.1487. [DOI] [PMC free article] [PubMed] [Google Scholar]



