Skip to main content

EDITORIAL article

Front. Oncol., 24 January 2024
Sec. Genitourinary Oncology
This article is part of the Research Topic Expert Opinions in Genitourinary Oncology View all 6 articles

Editorial: Expert opinions in genitourinary oncology

Galina G. Lagos,*Galina G. Lagos1,2*Wafik S. El-Deiry,,Wafik S. El-Deiry1,2,3Liang Cheng,*Liang Cheng2,3*
  • 1Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital and Brown University, Providence, RI, United States
  • 2Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI, United States
  • 3Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Providence, RI, United States

Editorial on the Research Topic
Expert opinions in genitourinary oncology

In the last decade we have seen an explosion in the development of new treatment options, diagnostic tools, and prognostic and predictive biomarkers for patients with genitourinary malignancies. As we learn to use these new tools, we face the challenge of appropriately identifying patients with high-risk disease that will benefit from our most aggressive treatment modalities and those with low-risk disease who are better served with more conservative management to avoid unnecessary toxicities. The articles in this Research Topic focus on approaches to stratifying patients with genitourinary malignancies in an effort to get the right treatment to the right patient at the right time.

The treatment landscape in prostate cancer is rapidly changing and we are fortunate to have a plethora of options for patients with metastatic hormone sensitive prostate cancer (mHSPC). Sigurdson et al have concisely and effectively summarized the standards of care for mHSPC as of 2020, which included androgen deprivation therapy (ADT) combined with either an androgen receptor signaling inhibitors (ARSI) or docetaxel. Since the publication of their article, two triplet therapy regimens have demonstrated superiority over ADT+docetaxel in the frontline setting. The PEACE-1 trial showed the efficacy of abiraterone/prednisone plus ADT and docetaxel (1) and the ARASENS trial established the role for darolutamide plus ADT and docetaxel (2). An unanswered question remains whether the addition of docetaxel has an added benefit over ADT+ARSI alone.

Furthermore, we have limited data to identify which patients are most appropriate for treatment intensification. In the CHAARTED trial, patients with high volume disease (HVD) were the subgroup of patients that benefited from the addition of docetaxel (3). In subgroup analysis from PEACE-1, benefit from triplet therapy was only observed for HVD patients, although in ARASENS benefit was seen across all subgroups (4). Another consideration is how we define high volume/high risk disease as we integrate PSMA-PET imaging in practice, given that prior trials used conventional imaging. Prospective trials using novel imaging modalities and biomarkers will be needed to better define the most appropriate frontline treatment for patients with mHSPC (Rosinha et al.).

In the metastatic castrate resistant prostate cancer (mCRPC) space, individualizing treatment based on molecular biomarkers has advanced patient care. Pathogenic variants in homologous recombination repair genes are identified in 20-30% of patients with mCRPC. These mutations confer sensitivity to PARP inhibitors. Olaparib was the first approved PARPi in mCRPC after progression on a prior ARSI based on the PROfound trial (5). Although the FDA granted approval for olaparib for multiple HRR gene variants, the benefit was most pronounced in patients with BRCA2 mutations, which has subsequently been seen across multiple PARPi trials. In the last year, on the basis of the PROpel, MAGNITUDE, and TALAPRO-2 studies, olaparib+abiraterone/prednisone, niraparib +abiraterone/prednisone, and talazoparib+enzalutamide, respectively, were all approved as first line regimens for mCRPC (68). In this setting, olaparib and niraparib were only approved in BRCA variants, although talazoparib has a broader approval for HRR mutations. Interestingly, in the TALAPRO-2 and PROpel studies, benefit was also seen in those without HRR mutations, although this benefit was less pronounced.

Another subgroup of patients where a biomarker-based approach has been valuable are the roughly 3% of prostate cancer patients with a microsatellite instability-high/mismatch repair deficient (MSI-H/MMRd) phenotype (9). In all-comers, outcomes with immune checkpoint inhibitors (ICI) in mCRPC have been disappointing across multiple trials including single agent and combination approaches. In a small series of patients with MSI-H/MMRd the response rate to checkpoint inhibitors are in the 45-53% range (9, 10). Based on the available data, pembrolizumab is now approved for this subset of patients, as well as those with high-TMB.

The question of treatment escalation or de-escalation is also relevant in the approach to earlier stages of prostate cancer. Champion et al describe the use of PSMA-PET in tailoring radiation approaches in post-prostatectomy patients. A third to one half of patients who undergo prostatectomy develop biochemical relapse. PSMA-PET scans have quickly become the preferred imaging modality to identify prostate cancer relapse in these patients. Given the novelty of this technology, most of the data regarding dose adjustment based on imaging findings is retrospective in nature. Based on the available data, PSMA-PET alone should not be used to guide radiation strategies but prospective studies are underway to help address this question (i.e. NCT04557501, NCT05067660, NCT05022914, NCT04794777). For the future, other biomarkers such as genomic classifiers and artificial intelligence-based tools may be incorporated to personalize treatment decisions.

Burke addresses the debate over our classification of low grade Gleason score 6 prostate cancers. Although some have argued to reclassify Gleason score 6 tumors as noncancer given their low mortality risk, Dr. Burke asserts that a severity prognostic score should not be used to deny the existence of a diagnosis. Despite a shared histology, not all Gleason 6 tumors are equal and over 50% of men with low-risk prostate cancer eventually receive cancer directed treatment. This speaks to the need for more granular prognostic biomarkers that can help provide reassurance to patients that will likely never need treatment and to prepare patients who may benefit from treatment down the line.

Beyond prostate cancer, there has been significant investment in developing better prognostic and predictive biomarkers to guide bladder cancer treatment. In muscle invasive bladder cancer (MIBC), the standard treatment for eligible patients is neoadjuvant chemotherapy followed by radical cystectomy. Given the morbidity and mortality risks, organ-sparing approaches are an attractive alternative. Trimodality therapy (TMT) using maximal transurethral resection of bladder tumor (TURBT) followed by chemotherapy with radiation is an option for patients who decline or are unfit for cystectomy. In current practice we use clinical factors including patient and tumor characteristics to identify candidates for TMT. For carefully selected patients, outcomes may be similar between TMT and radical cystectomy (11). Currently there are no validated biomarkers to select patients for TMT but areas of active investigation include molecular subtypes based on transcriptomic profiles, gene expression signatures of immune activation, and DNA damage response mutations (12). Ferro et al. describe pre-treatment lymphocyte-to-monocyte ratio (LMR) as another promising biomarker to identify patients at highest risk for progression which may guide therapy decisions.

It has also been observed that a subset of patients achieves long term bladder intact disease-free survival after TURBT plus systemic (13, 14). There are several completed trials and some underway to evaluate the role of TURBT plus systemic therapy as a definitive treatment option (1517). Some of these trials use biomarkers, including the presence of functionally deleterious DNA damage response gene alterations, which are associated with improved response to cisplatin-based therapies, to select appropriate candidates for observation after completion of chemotherapy. They also use stringent criteria to select patients who may avoid cystectomy, including a negative restaging TURBT, negative urine cytology, and no evidence of disease on MRI. Translational analyses from these studies will help define genomic, immunologic, and radiologic biomarkers to optimize patient selection for treatment approaches.

The newly evolving standard of care systemic therapy from metastatic bladder cancer is also likely to make its way in the earlier disease stages and broaden treatment options. As of this year the combination of enfortumab vedotin, an antibody drug conjugate targeting Nectin-4, with pembrolizumab, a PD-1 inhibitor, has been approved as an alternative to platinum based chemotherapy, which was been the frontline standard for decades. The phase III EV-302 trial demonstrated the superiority of this combination, which nearly doubled median OS and PFS relative to platinum based chemotherapy. Importantly, this effect was observed across pre-defined subgroups including cisplatin eligibility and PD-L1 expression (18). EV-304 is an ongoing trial exploring this combination in the neoadjuvant setting for muscle invasive disease with results eagerly awaited.

As therapeutic options for patients with genitourinary malignancies continue to expand, we must continue our efforts to tailor the right treatments for individual patients. Current research endeavors highlight evolving landscape of clinical management of genitourinary malignancies and the innovative approaches in development that will continue to push the field forward in coming years.

Author contributions

GL: Writing – original draft, Writing – review & editing. WE-D: Writing – original draft, Writing – review & editing. LC: Writing – original draft, Writing – review & editing.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Fizazi K, Foulon S, Carles J, Roubaud G, McDermott R, Fléchon A, et al. Abiraterone plus prednisone added to androgen deprivation therapy and docetaxel in de novo metastatic castration-sensitive prostate cancer (PEACE-1): a multicentre, open-label, randomised, phase 3 study with a 2 × 2 factorial design. Lancet (2022) 399(10336):1695–707. doi: 10.1016/S0140-6736(22)00367-1

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Smith MR, Hussain M, Saad F, Fizazi K, Sternberg CN, Crawford ED, et al. Darolutamide and survival in metastatic, hormone-sensitive prostate cancer. New Engl J Med (2022) 386(12):1132–42. doi: 10.1056/NEJMoa2119115

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Sweeney CJ, Chen YH, Carducci M, Liu G, Jarrard DF, Eisenberger M, et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. New Engl J Med (2015) 373(8):737–46. doi: 10.1056/NEJMoa1503747

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Hussain M, Tombal B, Saad F, Fizazi K, Sternberg CN, Crawford ED, et al. Darolutamide plus androgen-deprivation therapy and docetaxel in metastatic hormone-sensitive prostate cancer by disease volume and risk subgroups in the phase III ARASENS trial. JCO (2023) 41(20):3595–607. doi: 10.1200/JCO.23.00041

CrossRef Full Text | Google Scholar

5. de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Olaparib for metastatic castration-resistant prostate cancer. New Engl J Med (2020) 382(22):2091–102. doi: 10.1056/NEJMoa1911440

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Clarke NW, Armstrong AJ, Thiery-Vuillemin A, Oya M, Shore N, Loredo E, et al. Abiraterone and olaparib for metastatic castration-resistant prostate cancer. NEJM Evidence (2022) 1(9):EVIDoa2200043. doi: 10.1056/EVIDoa2200043

CrossRef Full Text | Google Scholar

7. Chi KN, Rathkopf D, Smith MR, Efstathiou E, Attard G, Olmos D, et al. Niraparib and abiraterone acetate for metastatic castration-resistant prostate cancer. J Clin Oncol (2023) 41(18):3339–51. doi: 10.1200/JCO.22.01649

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Agarwal N, Azad AA, Carles J, Fay AP, Matsubara N, Heinrich D, et al. Talazoparib plus enzalutamide in men with first-line metastatic castration-resistant prostate cancer (TALAPRO-2): a randomised, placebo-controlled, phase 3 trial. Lancet (2023) 402(10398):291–303. doi: 10.1016/S0140-6736(23)01055-3

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, et al. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol (2019) 5(4):471–8. doi: 10.1001/jamaoncol.2018.5801

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Graham LS, Montgomery B, Cheng HH, Yu EY, Nelson PS, Pritchard C, et al. Mismatch repair deficiency in metastatic prostate cancer: Response to PD-1 blockade and standard therapies. PloS One (2020) 15(5):e0233260. doi: 10.1371/journal.pone.0233260

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Zlotta AR, Ballas LK, Niemierko A, Lajkosz K, Kuk C, Miranda G, et al. Radical cystectomy versus trimodality therapy for muscle-invasive bladder cancer: a multi-institutional propensity score matched and weighted analysis. Lancet Oncol (2023) 24(6):669–81. doi: 10.1016/S1470-2045(23)00170-5

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Solanki AA, Venkatesulu BP, Efstathiou JA. Will the use of biomarkers improve bladder cancer radiotherapy delivery? Clin Oncol (2021) 33(6):e264–73. doi: 10.1016/j.clon.2021.03.017

CrossRef Full Text | Google Scholar

13. Meyer A, Ghandour R, Bergman A, Castaneda C, Wosnitzer M, Hruby G, et al. The natural history of clinically complete responders to neoadjuvant chemotherapy for urothelial carcinoma of the bladder. J Urol (2014) 192(3):696–701. doi: 10.1016/j.juro.2014.03.078

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Moran GW, Li G, Robins DJ, Matulay JT, McKiernan JM, Anderson CB. Systematic review and meta-analysis on the efficacy of chemotherapy with transurethral resection of bladder tumors as definitive therapy for muscle invasive bladder cancer. Bladder Cancer (2017) 3(4):245–58. doi: 10.3233/BLC-170134

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Galsky MD, Daneshmand S, Izadmehr S, Gonzalez-Kozlova E, Chan KG, Lewis S, et al. Gemcitabine and cisplatin plus nivolumab as organ-sparing treatment for muscle-invasive bladder cancer: a phase 2 trial. Nat Med (2023) 29(11):2825–34. doi: 10.1038/s41591-023-02568-1

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Geynisman DM, Abbosh P, Ross EA, Zibelman MR, Ghatalia P, Anari F, et al. A phase II trial of risk-enabled therapy after initiating neoadjuvant chemotherapy for bladder cancer (RETAIN). JCO (2023) 41(6_suppl):438–8. doi: 10.1200/JCO.2023.41.6_suppl.438

CrossRef Full Text | Google Scholar

17. Iyer G, Ballman KV, Atherton PJ, Murray K, Kwok Y, Steen PD, et al. A phase II study of gemcitabine plus cisplatin chemotherapy in patients with muscle-invasive bladder cancer with bladder preservation for those patients whose tumors harbor deleterious DNA damage response (DDR) gene alterations (Alliance A031701). JCO (2022) 40(16_suppl):TPS4615–TPS4615. doi: 10.1200/JCO.2022.40.16_suppl.TPS4615

CrossRef Full Text | Google Scholar

18. Powles TB, Valderrama BP, Gupta S, Bedke J, Kikuchi E, Hoffman-Censits J, et al. LBA6 EV-302/KEYNOTE-A39: Open-label, randomized phase III study of enfortumab vedotin in combination with pembrolizumab (EV+P) vs chemotherapy (Chemo) in previously untreated locally advanced metastatic urothelial carcinoma (la/mUC). Ann Oncol (2023) 34:S1340. doi: 10.1016/j.annonc.2023.10.106

CrossRef Full Text | Google Scholar

Keywords: precision oncology, clinical management, genitourinary cancer/malignancies, prognosis, prostate cancer, bladder cancer, kidney cancer

Citation: Lagos GG, El-Deiry WS and Cheng L (2024) Editorial: Expert opinions in genitourinary oncology. Front. Oncol. 13:1360223. doi: 10.3389/fonc.2023.1360223

Received: 22 December 2023; Accepted: 27 December 2023;
Published: 24 January 2024.

Edited and Reviewed by:

Ronald M Bukowski, Cleveland Clinic, United States

Copyright © 2024 Lagos, El-Deiry and Cheng. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Galina G. Lagos, galina_lagos@brown.edu; Liang Cheng, liang_cheng@yahoo.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.