Abstract
Purpose of review
In the present review, we summarize the recent developments in the management of germ cell tumors (GCTs).
Recent findings
Treatment-related acute and late-onset toxicity remains a key challenge in the management of GCTs. Recent data show that patients with large retroperitoneal lymph node metastases are at increased risk of venous thromboembolism and may benefit from prophylactic anticoagulation. Predictive models have been developed to identify patients with residual retroperitoneal lymph node masses that are more likely to benefit from surgical resection. However, their clinical use remains hampered by relatively low accuracy. There are currently multiple conventional-dose chemotherapy (CDCT) options for salvage therapy in patients with refractory or recurrent disease. In addition, more efficacious high-dose chemotherapy (HDCT) regimens continue to be developed. The role of salvage CDCT versus HDCT is currently being prospectively investigated. Finally, intratumoral heterogeneity is a common finding in cancer and an obvious observation in GCTs. Despite intratumoral heterogeneity, recent studies on nonseminomatous GCT have identified distinct histological subgroups and a potentially lethal clinical phenotype. Importantly, comprehensive molecular profiling so far has not elucidated the biologic basis or the clinical underpinnings of intratumoral heterogeneity in GCTs.
Summary
Remaining challenges to be addressed include minimizing therapeutic toxicity, and improving outcomes in patients with refractory/recurrent GCTs or malignant transformation of teratomas.
Keywords: Germ cell tumors, non-seminomas, seminomas, testicular cancer
INTRODUCTION
In the United States, an estimated 8720 new cases of testicular carcinoma will be diagnosed and only 380 will die of this disease in 2016 [1]. This excellent survival rate was achieved through a series of advances in cisplatin-based chemotherapy regimens over the past 50 years [2, 3]. However, for unclear reasons, the incidence of testicular cancer has been steadily increasing in the United States and Europe [4]. Given this rising incidence, and the greater than 35 years-of-life lost per patient [5], it is imperative to elucidate and target the mechanisms of chemotherapy resistance and cancer relapse in the small fraction of patients who succumb to their cancer. Germ cell tumors (GCTs) account for greater than 95% of testicular carcinomas [6]. Furthermore, a small proportion of GCTs originate outside the gonads, mainly in the mediastinum, with an incidence of 1.9–3.4 per 1 million [7]. There is substantial variability in the treatment response of these extragonadal GCTs, and particularly those of mediastinal origin are characterized by inferior clinical outcomes [8]. Therefore, a key remaining challenge is to understand the biologic differences between testicular and extragonadal GCTs, and improve the efficacy of our therapeutic approach.
Given the excellent outcomes in patients with testicular GCTs, particularly those with early-stage disease, the major challenge in these cases is how to reduce treatment-related morbidity without compromising clinical efficacy. In addition, since most patients will survive after a diagnosis of testicular GCT, it is imperative to understand and reduce the long-term risks of therapy. The present review summarizes the most recent advances in the management of GCTs (Table 1).
Table 1.
First author | Journal Year [Reference] | Highlights |
---|---|---|
Treatment-related morbidity | ||
Hauptmann | Br J Cancer 2016 [11] | Increased pancreatic cancer risk after radiation therapy |
Hashibe | J Cancer Surviv 2016 [16] | Late therapy-related toxicity: obesity |
Dusaud | J Surg Oncol 2016 [19] | Predicting residual masses with only necrosis |
Punjani | Can Urol Assoc J 2016 [23] | Predicting residual masses with only necrosis |
Mano | J Urol 2016 [24] | Predicting residual masses with only necrosis |
Gizzi | Eur J Cancer 2016 [27] | Risk of venous thromboembolism |
Ranganath | J Clin Oncol 2016 [29] | Pulmonary complications from bleomycin |
Salvage therapy | ||
Seidel | Urol Oncol 2016 [38] | Gemcitabine, oxaliplatin, paclitaxel: CR 13% |
Feldman | Clin Genitourin Cancer 2015 [40] | TI-CE followed by HDCT + SCT |
Nieto | Ann Oncol 2015 [41] | Bevacizumab/HDCT + SCT |
Giannatempo | J Urol 2016 [64] | Treatment of teratoma with somatic transformation |
Adra | J Clin Oncol 2016 [39] | HDCT+SCT experience in a large referral center |
Genomic and histological analyses | ||
Litchfield | Nat Commun 2015 [42] | Mutational spectrum from whole-exome sequencing |
Bagrodia | J Clin Oncol 2016 [43] | Cisplatin resistance and TP53/MDM2 defects |
Taylor-Weiner | Nature 2016 [44] | Genomic convergence, mitochondrial priming, GCT evolution |
Tu | Cancer 2016 [54] | Intratumoral heterogeneity, differentiation, and lethal subtype |
Bilen | Oncotarget 2016 [65] | Intratumoral heterogeneity and chemo-resistance |
Novel therapeutic targets | ||
Shah | Cancer Immunol Res 2016 [58] | Case report: anti-PD-1 therapy |
Necchi | Clin Genitourin Cancer 2016 [61] | Brentuximab vedotin targeting CD-30 expressing GCTs |
Abbreviations: HDCT, high-dose chemotherapy; SCT stem-cell transplantation; CR, complete response.
STRATEGIES TO MINIMIZE TREATMENT-RELATED MORBIDITY
Therapeutic chemotherapy and radiotherapy are recognized risk factors for secondary leukemia, lymphoma, as well as cancers of the bladder, colon, pancreas, kidney, stomach, thyroid, and soft tissue [9–12]. Management decisions should therefore aim to minimize exposure to these treatment modalities without compromising outcomes. In addition, survivors should be regularly monitored for the multiple other potential late-toxicities of GCT therapy such as cardiovascular disease, metabolic syndrome, hypogonadism, osteoporosis, and depression [13–15]. A recent population-based cohort study found that obesity prior to GCT diagnosis is associated with late therapy-related toxicity [16] and therefore these patients may potentially benefit from less intensive treatment modalities, more comprehensive lifestyle modifications, and closer long-term monitoring.
Surgical resection of residual retroperitoneal and/or lung masses is often required in patients with advanced GCTs. Retroperitoneal lymph node dissection (RPLND) of residual masses in patients with non-seminomatous germ cell tumors (NSGCTs) can reveal mature teratomas, necrotic tissue, or residual viable NSGCTs in 40%, 50%, and 10% of cases respectively [17]. Therefore, this potentially very complex and morbid procedure [18] could be avoided in approximately half of these patients if we could predict which residual masses will only contain necrosis [19]. Towards this goal, predictive models have become available [20, 21] but their clinical use remains limited by low accuracy [22], although this may be improved by minor modifications [23]. Furthermore, a recent retrospective analysis suggested that disease recurrence can occur, albeit infrequently, even in cases where only residual fibrosis/necrosis is found following RPLND [24]. Patients who have been heavily pretreated with multiple cycles of chemotherapy that included high-dose chemotherapy followed by autologous SCT are less likely to harbor necrosis in the RPLND specimens, whereas teratomas or viable cancer can be found in up to 72% of such cases [25].
Retrospective data suggest that patients with large retroperitoneal lymph node metastases (>5 cm in maximal axial diameter) of GCTs are at increased risk of venous thromboembolism (VTE), possibly due to decreased venous drainage [26]. Elevated lactic dehydrogenase (LDH) has also been found to be associated with increased VTE risk [27]. Such patients may benefit from preventive anticoagulation with low-molecular-weight heparin.
Patients with primary mediastinal NSGCTs may be at increased risk for pulmonary complications, particularly in the postoperative setting. For this reason, some experts prefer to treat these patients with VIP (etoposide, ifosfamide, cisplatin) rather than BEP (bleomycin, etoposide, cisplatin) in order to avoid potential bleomycin-induced toxicity [28]. A recent retrospective analysis of patients with primary mediastinal GCTs treated with postchemotherapy surgery at Indiana University suggested that BEP is associated with higher risk for serious pulmonary complications [29]. However, different time frames and patient selection could have biased these results. A cautionary note: if bleomycin-based regimens do provide survival benefit [30–32], avoidance of bleomycin could be a detriment to the care of certain patients with GCTs.
SALVAGE THERAPY FOR REFRACTORY OR RECURRENT GCTs
The optimal salvage strategy for patients with refractory or relapsed GCTs remains to be elucidated. Potential options include second line conventional-dose chemotherapy (CDCT) or high-dose chemotherapy (HDCT) followed by autologous stem-cell transplantation (SCT) [33]. Conventional-dose salvage chemotherapy regimens include VIP [34], TIP (paclitaxel, ifosfamide, cisplatin) [35], and VeIP (vinblastine, ifosfamide, cisplatin) [36]. None of these regimens have been compared head-to-head in this setting. However, TIP has emerged as the most commonly used CDCT, in large part because it has demonstrated a 63% durable remission rate [35] compared with 14.6% seen with VIP [34], and 23.7% with VeIP [36], although this difference may be inflated because the TIP trial did not include patients with primary mediastinal NSGCTs or with incomplete response to first-line therapy [35]. Oxaliplatin is a platinum-based chemotherapeutic agent with a different spectrum of antineoplastic activity compared with cisplatin [37]. For this reason, oxaliplatin is often used in patients with GCTs that are refractory to cisplatin-containing regimens. A recent data registry analysis showed that GOP (gemcitabine, oxaliplatin, paclitaxel) with or without additional tumor resection can achieve a complete remission rate of 13.1% when given outside of a clinical trial setting to patients refractory to cisplatin-based chemotherapy who received a median of 3 prior treatments, including HDCT with autologous stem cell transplant in 76% of cases [38].
Autologous SCT allows the administration of chemotherapy combinations at doses that would otherwise be fatal to patients. GCT is a highly chemosensitive malignancy occurring most often in young patients with few comorbidities who can tolerate the toxicities associated with autologous SCT relatively better than other patient populations. HDCT followed by peripheral-blood autologous SCT can achieve a 2-year progression-free survival of 63% when given as second-line therapy, and 49% when given as third-line or later [39]. The TI-CE regimen consists of paclitaxel and ifosfamide induction for up to 2 cycles followed by high-dose carboplatin and etoposide with autologous SCT every 21–28 days for 3 cycles. The efficacy of this approach was demonstrated in a phase I/II trial that reported a 47% 5-year disease-free survival and 52% overall survival [40]. The TIGER trial (NCT02375204 at www.clinicaltrials.gov) is an ongoing multicenter, open-label phase III study comparing second-line TI-CE vs TIP in patients with relapsed or refractory GCTs. This trial will help determine which patients can benefit more from first salvage therapy with HDCT versus CDCT. Another recent single-arm phase II trial used a rationally designed HDCT combination of bevacizumab, gemcitabine, docetaxel, melphalan, and carboplatin (cycle 1) followed by bevacizumab, ifosfamide, etoposide, and carboplatin (cycle 2) [41]. This strategy exceeded the reported outcomes of prior HDCT regimens, particularly in patients with poor-risk features per the Beyer prognostic model.
INSIGHTS GAINED BY GENOMIC AND HISTOLOGICAL ANALYSES
Although GCTs are currently classified according to their histopathological appearance and anatomical site of origin, more refined taxonomies integrating genomic, epigenomic, transcriptomic, and proteomic data may allow more accurate prognostication and management. Such comprehensive profiling has been undertaken by The Cancer Genome Atlas project (https://cancergenome.nih.gov/) and the results are publicly available for investigators to use in generating or validating hypotheses based on the observed genomic and biological patterns. Whole exome sequencing of testicular GCTs has revealed a relatively low frequency of somatic point mutations (average mutation rate of 0.5–0.9 per Mb) which supports the embryological origin of this malignancy [42–44]. Conversely, chromosome arm-level copy number gains, particularly the presence of an isochromosome of the short arm of chromosome 12 (i12p), are significantly more frequent events [42, 44]. Most GCTs are hyperploid based on cytogenetic studies [45]. The exact role of the genes amplified within i12p remains to be elucidated.
Alterations in KIT and KRAS genes are the most frequent non-synonymous mutations found in GCTs, suggesting that these oncogenes may play an important role in the pathogenesis and growth of these tumors [42, 43, 46]. Nevertheless, a clinical trial of the KIT inhibitor imatinib did not demonstrate antitumor activity against platinum-refractory GCTs positive for KIT expression by immunohistochemistry [47]. It is possible that targeted inhibition of the KIT pathway may be necessary but not sufficient on its own to achieve lethality in these tumors. Furthermore, in contrast to imatinib-sensitive malignancies such as gastrointestinal stromal tumors, KIT mutations in GCTs are mainly localized on exon 17 and are thus more likely to confer resistance to imatinib [43].
Bagrodia et al. [43] showed that 25 of 104 (24%) cisplatin-resistant tumors had genetic defects within the TP53/MDM2 pathway compared with 2 of 76 (2.6%) of the cisplatin sensitive tumors (P<0.001). Of note, patients in the cisplatin-resistant cohort had a low death rate (23.1%) and it is unclear whether these patients who succumbed to their disease following salvage platinum-based treatments (such as TIP and high-dose carboplatin) were the ones who had TP53/MDM2 defects or these alterations only predict resistance to first-line cisplatin-based chemotherapy.
Similarly, Taylor-Weiner et al. [44] demonstrated genomic convergence, but did not discern any histological subgroups with prognostic significance, transformation potential, or lethal phenotypes. Because microdissection was not performed, it was unclear whether heterogeneous tumor populations in a mixed GCT (containing embryonal carcinoma, yolk sac tumor, choriocarcinoma, seminoma, and/or teratoma) had disparate mitochondrial priming capabilities [48, 49]. Although the authors showed loss of pluripotent gene expression, NANOG and POU5f1, this would not be unexpected in a differentiated tumor such as teratoma (i.e., GCT evolution). Importantly, intact TP53 and i(12p) were likely to be present in both chemo-sensitive embryonal carcinoma and chemo-resistant teratoma if microdissection had been performed to delineate diverse histological components [50, 51].
GCT is a prototype stem-cell tumor capable of differentiating into multiple lineages and phenotypes. It is an ideal tumor model to test the stem-cell theory of cancer [52] and for the study of intratumoral heterogeneity in which pluripotent cancer cells (e.g., embryonal carcinoma) are intermingled with differentiated cancer cells (e.g., teratoma), and where different tumor components exert or exhibit differential mitochondrial priming despite similar genetic makeup due to a common clonal origin [53]. Tu et al. [54] identified tumor subgroups based on developmental pathways and histological makeups that provided prognostic significance, predicted transformation risk, and revealed lethal phenotypes among GCTs. Their data suggest that embryonic origins may be both biologically and clinically relevant for the purpose of subgrouping GCTs.
NOVEL THERAPEUTIC TARGETS
Therapies targeting immune checkpoint molecules, such as the cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 (PD-1), have shown excellent efficacy against a variety of malignancies and have thus become powerful new additions to the oncologist’s toolbox [55]. PD-L1 is the PD-1 ligand that is often aberrantly expressed on cancer cells resulting in suppression of anti-tumor immunity via the PD-1 signaling pathway. PD-L1 has been found to be expressed in 73–76% of seminonas and 64–89% of NSGCTs at significantly higher levels compared with untransformed testicular tissue [56, 57]. This indicates that agents targeting the PD-1/PD-L1 interaction may be efficacious against GCTs. A recent case report described a patient with advanced embryonal carcinoma who had a rapid clinical response following one dose of first-line nivolumab given erroneously after being misdiagnosed with melanoma [58]. Subsequent analysis of The Cancer Genome Atlas GCT cohort by the authors revealed that almost half of GCT tissues demonstrate evidence of a T-cell-inflamed tumor microenvironment [58]. It is therefore conceivable that therapies targeting the PD-1/PD-L1 pathway may activate and/or attract anti-tumor effector T cells in GCTs. These could be further potentiated by combination with other agents such as anti-CTLA4 antibodies.
The cytokine receptor CD30 is expressed in embryonal carcinomas [59] and some seminomas [60]. Accordingly, 9 patients with CD30-expressing cisplatin-resistant GCTs were treated with the CD30-targeted monoclonal antibody brentuximab as salvage monotherapy in a small phase II trial [61]. Only 1 patient showed a complete response, another one had a partial response, and 2 achieved stable disease [61]. The tumors quickly developed resistance to therapy, as evidence by increase of serum tumor markers [61]. Therefore, brentuximab is more likely to have a role in CD30-positive GCTs as part of combination regimens rather than monotherapy.
OTHER STRATEGIES TO IMPROVE OUTCOMES
Although GCTs are highly curable tumors in most cases, patients with very poor prognostic features can have 3-year survival rates as low as 6.1% [62]. It is clear that more individualized approaches should be used in these cases. A practical strategy to improve clinical outcome is to identify potentially vulnerable patient subgroups and lethal tumor phenotypes based on pertinent clinical observations and a valid scientific rationale so that an integrated, and perhaps multimodal, targeted approach can be applied in a timely manner. For example, such patients may be predisposed to undergo somatic transformation, and therefore may benefit more from RPLND rather than active surveillance.
For patients whose primary GCTs contain yolk sac tumor and seminoma are at higher risk of developing somatic transformation within their metastatic lesions after chemotherapy, and for death [54]. Of interest, patients whose primary tumors contained the same components along with somatic transformation did not experience a worse outcome [54]. This is consistent with reports indicating that teratomas with malignant transformation confer a much worse prognosis than other GCTs, if surgical resection is not feasible [63, 64].
Similarly, certain patients with clinical stage I GCT whose primary testicular tumor contains yolk sac tumor and seminoma or mixed teratoma (in contrast to pure embryonal carcinoma) may be prime candidates for RPLND, because their residual tumor after chemotherapy tends to contain teratoma and is at increased risk to undergo somatic transformation [54, 65]. These patients may not be ideal candidates for active surveillance, because when they develop recurrent disease they appear to be at higher risk of dying from refractory disease [65]
Therefore, a key to improve outcomes is recognition and awareness of a potentially lethal subgroup of testicular GCT. When an indolent but refractory GCT becomes systemic, the window of opportunity towards a cure may be closed. We propose that early diagnosis and prompt systemic treatment using chemotherapy and localized therapy comprising surgery will improve the clinical outcome and enhance the cure rate of this potentially lethal GCT entity.
CONCLUSION
Significant advances made in the past 50 years have achieved high cure rates even in patients with very advanced germ cell tumors (GCTs). Nevertheless, key remaining challenges include minimizing treatment-related acute and long-term toxicities, optimizing the selection of salvage therapies for refractory or recurrent GCTs, and testing novel agents for the treatment of patients who have progressed on the current standard cytotoxic agents (Table 1). The current management approaches can impose a substantial burden of unnecessary treatment-related toxicity in some patients who otherwise respond well to therapy, and by the same token can fail others who ultimately succumb to their disease. To address these issues, we must continue to individualize our management decisions, better understand when to deliver new agents, and how to layer in or supplant chemotherapy. Emerging technologies have enabled the comprehensive molecular profiling of GCTs, leading to new biological insights. However, advanced technology, arcane bioinformatics, and intricate experiments will not help us address unsettled questions, devise precise prognostication, and identify plausible therapeutic targets to improve clinical outcomes without the benefit of pertinent clinical observations and the backing of a veritable scientific hypothesis [66, 67].
KEY POINTS.
Minimizing acute and late-onset toxicity is a key challenge in the management of germ cell tumors.
The role and optimal timing of high-dose chemotherapy followed by stem-cell transplantation remains to be determined and is currently being prospectively investigated.
Intratumoral heterogeneity can be traced to developmental pathways and separated by histological subgroups with biologic, prognostic, and therapeutic implications.
The comprehensive molecular profiling of germ cell tumors may uncover potentially targetable biological pathways and processes.
Acknowledgments
Pavlos Msaouel is supported by the National Institutes of Health T32 CA009666 grant. This work was supported in part by the National Institutes of Health through MD Anderson’s Cancer Center Support Grant CA016672 (used the Clinical Trials Support Resource)
References
- 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7–30. doi: 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
- 2.Hanna N, Einhorn LH. Testicular cancer: a reflection on 50 years of discovery. J Clin Oncol. 2014;32:3085–92. doi: 10.1200/JCO.2014.56.0896. [DOI] [PubMed] [Google Scholar]
- 3.Hanna NH, Einhorn LH. Testicular cancer--discoveries and updates. N Engl J Med. 2014;371:2005–16. doi: 10.1056/NEJMra1407550. [DOI] [PubMed] [Google Scholar]
- 4.Nigam M, Aschebrook-Kilfoy B, Shikanov S, Eggener S. Increasing incidence of testicular cancer in the United States and Europe between 1992 and 2009. World J Urol. 2015;33:623–31. doi: 10.1007/s00345-014-1361-y. [DOI] [PubMed] [Google Scholar]
- 5.Li C, Ekwueme DU, Rim SH, Tangka FK. Years of potential life lost and productivity losses from male urogenital cancer deaths--United States, 2004. Urology. 2010;76:528–35. doi: 10.1016/j.urology.2010.04.030. [DOI] [PubMed] [Google Scholar]
- 6.Cheville JC. Classification and pathology of testicular germ cell and sex cord-stromal tumors. Urol Clin North Am. 1999;26:595–609. doi: 10.1016/s0094-0143(05)70201-9. [DOI] [PubMed] [Google Scholar]
- 7.Stang A, Trabert B, Wentzensen N, et al. Gonadal and extragonadal germ cell tumours in the United States, 1973–2007. Int J Androl. 2012;35:616–25. doi: 10.1111/j.1365-2605.2011.01245.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ko JJ, Asif T, Li H, et al. Disease characteristics and survival outcomes of extragonadal primary germ cell tumour in two Canadian tertiary cancer centres. Can Urol Assoc J. 2016;10:E165–E70. doi: 10.5489/cuaj.3357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Fung C, Fossa SD, Milano MT, et al. Solid tumors after chemotherapy or surgery for testicular nonseminoma: a population-based study. J Clin Oncol. 2013;31:3807–14. doi: 10.1200/JCO.2013.50.3409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Abouassaly R, Fossa SD, Giwercman A, et al. Sequelae of treatment in long-term survivors of testis cancer. Eur Urol. 2011;60:516–26. doi: 10.1016/j.eururo.2011.05.055. [DOI] [PubMed] [Google Scholar]
- 11.Hauptmann M, Borge Johannesen T, Gilbert ES, et al. Increased pancreatic cancer risk following radiotherapy for testicular cancer. Br J Cancer. 2016;115:901–8. doi: 10.1038/bjc.2016.272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Travis LB, Curtis RE, Storm H, et al. Risk of second malignant neoplasms among long-term survivors of testicular cancer. J Natl Cancer Inst. 1997;89:1429–39. doi: 10.1093/jnci/89.19.1429. [DOI] [PubMed] [Google Scholar]
- 13.Willemse PM, Burggraaf J, Hamdy NA, et al. Prevalence of the metabolic syndrome and cardiovascular disease risk in chemotherapy-treated testicular germ cell tumour survivors. Br J Cancer. 2013;109:60–7. doi: 10.1038/bjc.2013.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Haugnes HS, Wethal T, Aass N, et al. Cardiovascular risk factors and morbidity in long-term survivors of testicular cancer: a 20-year follow-up study. J Clin Oncol. 2010;28:4649–57. doi: 10.1200/JCO.2010.29.9362. [DOI] [PubMed] [Google Scholar]
- 15.Shinn EH, Swartz RJ, Thornton BB, et al. Testis cancer survivors’ health behaviors: comparison with age-matched relative and demographically matched population controls. J Clin Oncol. 2010;28:2274–9. doi: 10.1200/JCO.2009.23.9608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hashibe M, Abdelaziz S, Al-Temimi M, et al. Long-term health effects among testicular cancer survivors. J Cancer Surviv. 2016;10:1051–7. doi: 10.1007/s11764-016-0548-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Fox EP, Weathers TD, Williams SD, et al. Outcome analysis for patients with persistent nonteratomatous germ cell tumor in postchemotherapy retroperitoneal lymph node dissections. J Clin Oncol. 1993;11:1294–9. doi: 10.1200/JCO.1993.11.7.1294. [DOI] [PubMed] [Google Scholar]
- 18.Heidenreich A, Haidl F, Paffenholz P, et al. Surgical management of complex residual masses following systemic chemotherapy for metastatic testicular germ cell tumours. Ann Oncol. 2016 doi: 10.1093/annonc/mdw605. [DOI] [PubMed] [Google Scholar]
- 19.Dusaud M, Malavaud B, Bayoud Y, et al. Post-chemotherapy retroperitoneal teratoma in nonseminomatous germ cell tumors: Do predictive factors exist? Results from a national multicenter study. J Surg Oncol. 2016 doi: 10.1002/jso.24464. [DOI] [PubMed] [Google Scholar]
- 20.Albers P, Weissbach L, Krege S, et al. Prediction of necrosis after chemotherapy of advanced germ cell tumors: results of a prospective multicenter trial of the German Testicular Cancer Study Group. J Urol. 2004;171:1835–8. doi: 10.1097/01.ju.0000119121.36427.09. [DOI] [PubMed] [Google Scholar]
- 21.Vergouwe Y, Steyerberg EW, Foster RS, et al. Predicting retroperitoneal histology in postchemotherapy testicular germ cell cancer: a model update and multicentre validation with more than 1000 patients. Eur Urol. 2007;51:424–32. doi: 10.1016/j.eururo.2006.06.047. [DOI] [PubMed] [Google Scholar]
- 22.Heidenreich A. Residual tumour resection following inductive chemotherapy in advanced testicular cancer. Eur Urol. 2007;51:299–301. doi: 10.1016/j.eururo.2006.06.046. [DOI] [PubMed] [Google Scholar]
- 23•.Punjani N, Power N, Vanhie JJ, Winquist E. Validation of a prediction model for avoiding post-chemotherapy retroperitoneal lymphadenectomy in patients with metastatic nonseminomatous germ cell cancer. Can Urol Assoc J. 2016;10:260–3. doi: 10.5489/cuaj.3558. Latest modified version of a prediction model to determine which patients are more likely to benefit from retroperitoneal lymph node dissection of residual massses post-chemotherapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24•.Mano R, Becerra MF, Carver BS, et al. Clinical Outcome of Patients with Fibrosis/Necrosis at Post-Chemotherapy Retroperitoneal Lymph Node Dissection for Advanced Germ Cell Tumors. J Urol. 2016 doi: 10.1016/j.juro.2016.09.113. This report indicates that patients with residual post-chemotherapy fibrosis/necrosis may still relapse. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cary C, Pedrosa JA, Jacob J, et al. Outcomes of postchemotherapy retroperitoneal lymph node dissection following high-dose chemotherapy with stem cell transplantation. Cancer. 2015;121:4369–75. doi: 10.1002/cncr.29678. [DOI] [PubMed] [Google Scholar]
- 26••.Srikanthan A, Tran B, Beausoleil M, et al. Large retroperitoneal lymphadenopathy as a predictor of venous thromboembolism in patients with disseminated germ cell tumors treated with chemotherapy. J Clin Oncol. 2015;33:582–7. doi: 10.1200/JCO.2014.58.6537. Study showing that large retroperitoneal lymphadenopathy is associated with increased risk of venous thromboembolism. [DOI] [PubMed] [Google Scholar]
- 27.Gizzi M, Oberic L, Massard C, et al. Predicting and preventing thromboembolic events in patients receiving cisplatin-based chemotherapy for germ cell tumours. Eur J Cancer. 2016;69:151–7. doi: 10.1016/j.ejca.2016.10.003. [DOI] [PubMed] [Google Scholar]
- 28.Albany C, Einhorn LH. Extragonadal germ cell tumors: clinical presentation and management. Curr Opin Oncol. 2013;25:261–5. doi: 10.1097/CCO.0b013e32835f085d. [DOI] [PubMed] [Google Scholar]
- 29•.Ranganath P, Kesler KA, Einhorn LH. Perioperative Morbidity and Mortality Associated With Bleomycin in Primary Mediastinal Nonseminomatous Germ Cell Tumor. J Clin Oncol. 2016 doi: 10.1200/JCO.2016.69.8910. Retrospective analysis suggesting that patients with primary mediastinal nonseminomatous germ cell tumors may have less post-surgical pulmonary toxicity if previously treated with VIP compared with BEP. [DOI] [PubMed] [Google Scholar]
- 30.de Wit R, Stoter G, Kaye SB, et al. Importance of bleomycin in combination chemotherapy for good-prognosis testicular nonseminoma: a randomized study of the European Organization for Research and Treatment of Cancer Genitourinary Tract Cancer Cooperative Group. J Clin Oncol. 1997;15:1837–43. doi: 10.1200/JCO.1997.15.5.1837. [DOI] [PubMed] [Google Scholar]
- 31.Loehrer PJ, Sr, Johnson D, Elson P, et al. Importance of bleomycin in favorable-prognosis disseminated germ cell tumors: an Eastern Cooperative Oncology Group trial. J Clin Oncol. 1995;13:470–6. doi: 10.1200/JCO.1995.13.2.470. [DOI] [PubMed] [Google Scholar]
- 32.Levi JA, Raghavan D, Harvey V, et al. The importance of bleomycin in combination chemotherapy for good-prognosis germ cell carcinoma. Australasian Germ Cell Trial Group. J Clin Oncol. 1993;11:1300–5. doi: 10.1200/JCO.1993.11.7.1300. [DOI] [PubMed] [Google Scholar]
- 33.Rashdan S, Einhorn LH. Salvage Therapy for Patients With Germ Cell Tumor. J Oncol Pract. 2016;12:437–43. doi: 10.1200/JOP.2016.011411. [DOI] [PubMed] [Google Scholar]
- 34.Loehrer PJ, Sr, Einhorn LH, Williams SD. VP-16 plus ifosfamide plus cisplatin as salvage therapy in refractory germ cell cancer. J Clin Oncol. 1986;4:528–36. doi: 10.1200/JCO.1986.4.4.528. [DOI] [PubMed] [Google Scholar]
- 35.Kondagunta GV, Bacik J, Donadio A, et al. Combination of paclitaxel, ifosfamide, and cisplatin is an effective second-line therapy for patients with relapsed testicular germ cell tumors. J Clin Oncol. 2005;23:6549–55. doi: 10.1200/JCO.2005.19.638. [DOI] [PubMed] [Google Scholar]
- 36.Loehrer PJ, Sr, Gonin R, Nichols CR, et al. Vinblastine plus ifosfamide plus cisplatin as initial salvage therapy in recurrent germ cell tumor. J Clin Oncol. 1998;16:2500–4. doi: 10.1200/JCO.1998.16.7.2500. [DOI] [PubMed] [Google Scholar]
- 37.Perego P, Robert J. Oxaliplatin in the era of personalized medicine: from mechanistic studies to clinical efficacy. Cancer Chemother Pharmacol. 2016;77:5–18. doi: 10.1007/s00280-015-2901-x. [DOI] [PubMed] [Google Scholar]
- 38•.Seidel C, Oechsle K, Lorch A, et al. Efficacy and safety of gemcitabine, oxaliplatin, and paclitaxel in cisplatin-refractory germ cell cancer in routine care--Registry data from an outcomes research project of the German Testicular Cancer Study Group. Urol Oncol. 2016;34:167.e21–8. doi: 10.1016/j.urolonc.2015.11.007. GOP regimen. [DOI] [PubMed] [Google Scholar]
- 39••.Adra N, Abonour R, Althouse SK, et al. High-Dose Chemotherapy and Autologous Peripheral-Blood Stem-Cell Transplantation for Relapsed Metastatic Germ Cell Tumors: The Indiana University Experience. J Clin Oncol. 2016 doi: 10.1200/JCO.2016.69.5395. JCO2016695395 This paper reports the experience of a large referral center with salvage high-dose chemotherapy followed by stem-cell transplantation in patients with germ cell tumors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40••.Feldman DR, Glezerman I, Patil S, et al. Phase I/II Trial of Paclitaxel With Ifosfamide Followed by High-Dose Paclitaxel, Ifosfamide, and Carboplatin (TI-TIC) With Autologous Stem Cell Reinfusion for Salvage Treatment of Germ Cell Tumors. Clin Genitourin Cancer. 2015;13:453–60. doi: 10.1016/j.clgc.2015.05.003. This paper report the toxicity/efficacy characteristics of the TI-CE regimen. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41••.Nieto Y, Tu SM, Bassett R, et al. Bevacizumab/high-dose chemotherapy with autologous stem-cell transplant for poor-risk relapsed or refractory germ-cell tumors. Ann Oncol. 2015;26:2125–32. doi: 10.1093/annonc/mdv310. This paper reports the clinical outcomes of a rationally designed salvage high-dose chemotherapy regimen for germ cell tumors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42••.Litchfield K, Summersgill B, Yost S, et al. Whole-exome sequencing reveals the mutational spectrum of testicular germ cell tumours. Nat Commun. 2015;6:5973. doi: 10.1038/ncomms6973. This study used whole-exome sequencing to profile of testicular germ cell tumors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43••.Bagrodia A, Lee BH, Lee W, et al. Genetic Determinants of Cisplatin Resistance in Patients With Advanced Germ Cell Tumors. J Clin Oncol. 2016 doi: 10.1200/JCO.2016.68.7798. This study investigated the molecular alterations associated with cisplatin resistance in patients with germ cell tumors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44••.Taylor-Weiner A, Zack T, O’Donnell E, et al. Genomic evolution and chemoresistance in germ-cell tumours. Nature. 2016;540:114–8. doi: 10.1038/nature20596. This important study performed whole-exome and transcriptome profiling of testicular and primary mediastinal germ cell tumors and found that the increased chemosensitity of many of these tumors may be due to high mitochondrial priming for apoptosis in the setting of an intact TP53 pathway status. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Oosterhuis JW, Castedo SM, de Jong B, et al. Ploidy of primary germ cell tumors of the testis. Pathogenetic and clinical relevance. Lab Invest. 1989;60:14–21. [PubMed] [Google Scholar]
- 46.Cutcutache I, Suzuki Y, Tan IB, et al. Exome-wide Sequencing Shows Low Mutation Rates and Identifies Novel Mutated Genes in Seminomas. Eur Urol. 2015;68:77–83. doi: 10.1016/j.eururo.2014.12.040. [DOI] [PubMed] [Google Scholar]
- 47.Einhorn LH, Brames MJ, Heinrich MC, et al. Phase II study of imatinib mesylate in chemotherapy refractory germ cell tumors expressing KIT. Am J Clin Oncol. 2006;29:12–3. doi: 10.1097/01.coc.0000195086.47548.ef. [DOI] [PubMed] [Google Scholar]
- 48.Liu JC, Guan X, Ryan JA, et al. High mitochondrial priming sensitizes hESCs to DNA-damage-induced apoptosis. Cell Stem Cell. 2013;13:483–91. doi: 10.1016/j.stem.2013.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhou W, Choi M, Margineantu D, et al. HIF1alpha induced switch from bivalent to exclusively glycolytic metabolism during ESC-to-EpiSC/hESC transition. EMBO J. 2012;31:2103–16. doi: 10.1038/emboj.2012.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Jones TD, Wang M, Sung MT, et al. Clonal origin of metastatic testicular teratomas. Clin Cancer Res. 2006;12:5377–83. doi: 10.1158/1078-0432.CCR-06-0444. [DOI] [PubMed] [Google Scholar]
- 51.Kum JB, Ulbright TM, Williamson SR, et al. Molecular genetic evidence supporting the origin of somatic-type malignancy and teratoma from the same progenitor cell. Am J Surg Pathol. 2012;36:1849–56. doi: 10.1097/PAS.0b013e31826df1ab. [DOI] [PubMed] [Google Scholar]
- 52.Tu SM, Lin SH, Logothetis CJ. Stem-cell origin of metastasis and heterogeneity in solid tumours. Lancet Oncol. 2002;3:508–13. doi: 10.1016/s1470-2045(02)00820-3. [DOI] [PubMed] [Google Scholar]
- 53.Tu SM. Origin of cancers. Clinical perspectives and implications of a stem-cell theory of cancer. Cancer Treat Res. 2010;154:v–239. doi: 10.1007/978-1-4419-5968-3_1. [DOI] [PubMed] [Google Scholar]
- 54••.Tu SM, Bilen MA, Hess KR, et al. Intratumoral heterogeneity: Role of differentiation in a potentially lethal phenotype of testicular cancer. Cancer. 2016;122:1836–43. doi: 10.1002/cncr.29996. Research study showing that certain histological characteristics are associated with unfavorable prognosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61. doi: 10.1126/science.aaa8172. [DOI] [PubMed] [Google Scholar]
- 56•.Fankhauser CD, Curioni-Fontecedro A, Allmann V, et al. Frequent PD-L1 expression in testicular germ cell tumors. Br J Cancer. 2015;113:411–3. doi: 10.1038/bjc.2015.244. Evaluation of PD-L1 expression in testicular germ cell tumors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57•.Cierna Z, Mego M, Miskovska V, et al. Prognostic value of programmed-death-1 receptor (PD-1) and its ligand 1 (PD-L1) in testicular germ cell tumors. Ann Oncol. 2016;27:300–5. doi: 10.1093/annonc/mdv574. Evaluation of PD-1 and PD-L1 expression in testicular germ cell tumors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58•.Shah S, Ward JE, Bao R, et al. Clinical Response of a Patient to Anti-PD-1 Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors. Cancer Immunol Res. 2016;4:903–9. doi: 10.1158/2326-6066.CIR-16-0087. Case report of a patient with advanced embryonal carcinoma who experienced rapid improvement after one dose of first-line nivolumab. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Giannatempo P, Paolini B, Miceli R, et al. Persistent CD30 expression by embryonal carcinoma in the treatment time course: prognostic significance of a worthwhile target for personalized treatment. J Urol. 2013;190:1919–24. doi: 10.1016/j.juro.2013.04.057. [DOI] [PubMed] [Google Scholar]
- 60.Tickoo SK, Hutchinson B, Bacik J, et al. Testicular seminoma: a clinicopathologic and immunohistochemical study of 105 cases with special reference to seminomas with atypical features. Int J Surg Pathol. 2002;10:23–32. doi: 10.1177/106689690201000105. [DOI] [PubMed] [Google Scholar]
- 61.Necchi A, Anichini A, Raggi D, et al. Brentuximab Vedotin in CD30-Expressing Germ Cell Tumors After Chemotherapy Failure. Clin Genitourin Cancer. 2016;14:261–4. e4. doi: 10.1016/j.clgc.2016.03.020. [DOI] [PubMed] [Google Scholar]
- 62.International Prognostic Factors Study G. Lorch A, Beyer J, et al. Prognostic factors in patients with metastatic germ cell tumors who experienced treatment failure with cisplatin-based first-line chemotherapy. J Clin Oncol. 2010;28:4906–11. doi: 10.1200/JCO.2009.26.8128. [DOI] [PubMed] [Google Scholar]
- 63.Mikuz G, Colecchia M. Teratoma with somatic-type malignant components of the testis. A review and an update. Virchows Arch. 2012;461:27–32. doi: 10.1007/s00428-012-1251-x. [DOI] [PubMed] [Google Scholar]
- 64.Giannatempo P, Pond GR, Sonpavde G, et al. Treatment and Clinical Outcomes of Patients with Teratoma with Somatic-Type Malignant Transformation: An International Collaboration. J Urol. 2016;196:95–100. doi: 10.1016/j.juro.2015.12.082. [DOI] [PubMed] [Google Scholar]
- 65••.Bilen MA, Hess KR, Campbell MT, et al. Intratumoral heterogeneity and chemoresistance in nonseminomatous germ cell tumor of the testis. Oncotarget. 2016 doi: 10.18632/oncotarget.13380. Additional research showing that certain histological characteristics are associated with unfavorable prognosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tu SM, Bilen MA, Tannir NM. Personalised cancer care: promises and challenges of targeted therapy. J R Soc Med. 2016;109:98–105. doi: 10.1177/0141076816631154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Tu SM, Bilen MA, Tannir NM. The scientific method: pillar and pitfall of cancer research. Cancer Med. 2014;3:1035–7. doi: 10.1002/cam4.248. [DOI] [PMC free article] [PubMed] [Google Scholar]