351
|
Neuroendocrine and Aggressive-Variant Prostate Cancer. Cancers (Basel) 2020; 12:cancers12123792. [PMID: 33339136 PMCID: PMC7765615 DOI: 10.3390/cancers12123792] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 01/12/2023] Open
Abstract
In prostate cancer, neuroendocrine (NE) differentiation may rarely present de novo or more frequently arises following hormonal therapy in patients with castration-resistant prostate cancer (CRPC). Its distinct phenotype is characterized by an aggressive clinical course, lack of responsiveness to hormonal therapies and poor prognosis. Importantly, a subset of CRPC patients exhibits an aggressive-variant disease with very similar clinical and molecular characteristics to small-cell prostate cancer (SCPC) even though tumors do not have NE differentiation. This aggressive-variant prostate cancer (AVPC) also shares the sensitivity of SCPC to platinum-based chemotherapy albeit with short-lived clinical benefit. As optimal treatment strategies for AVPC remain elusive, currently ongoing research efforts aim to enhance our understanding of the biology of this disease entity and improve treatment outcomes for our patients. This review is an overview of our current knowledge on prostate cancer with NE differentiation and AVPC, with a focus on their clinical characteristics and management, including available as well as experimental therapeutic strategies.
Collapse
|
352
|
Smits M, Ekici K, Pamidimarri Naga S, van Oort IM, Sedelaar MJP, Schalken JA, Nagarajah J, Scheenen TWJ, Gerritsen WR, Fütterer JJ, Mehra N. Prior PSMA PET-CT Imaging and Hounsfield Unit Impact on Tumor Yield and Success of Molecular Analyses from Bone Biopsies in Metastatic Prostate Cancer. Cancers (Basel) 2020; 12:cancers12123756. [PMID: 33327413 PMCID: PMC7764855 DOI: 10.3390/cancers12123756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Prostate cancer is currently the fifth leading cause of death in men worldwide. To personalize and guide treatment in prostate cancer, identification of druggable genomic alterations is of major importance. Prostate cancer often metastasizes solely or predominantly to the bones, with molecular analyses on bone biopsies challenging due to technical difficulties to identify and obtain biopsies from high tumor cell containing locations. In our retrospective analysis, we showed a significantly higher success rate in patients where biopsy location was selected by a prior PSMA PET-CT compared to solely CT or MRI. CT-guided biopsies in locations with low Hounsfield units (HUs) and deviation of HUs were associated with a higher proportion of successful histological and molecular biopsies. Based on these results, we designed a simple prediction model for daily clinical practice to increase the success rate of bone biopsies for molecular analyses in prostate cancer to guide precision medicine. Abstract Developing and optimizing targeted therapies in metastatic castration-resistant prostate cancer (mCRPC) necessitates molecular characterization. Obtaining sufficient tumor material for molecular characterization has been challenging. We aimed to identify clinical and imaging variables of imaging-guided bone biopsies in metastatic prostate cancer patients that associate with tumor yield and success in obtaining molecular results, and to design a predictive model: Clinical and imaging data were collected retrospectively from patients with prostate cancer who underwent a bone biopsy for histological and molecular characterization. Clinical characteristics, imaging modalities and imaging variables, were associated with successful biopsy results. In our study, we included a total of 110 bone biopsies. Histological conformation was possible in 84 of all biopsies, of which, in 73 of the 84, successful molecular characterization was performed. Prior use of PSMA PET-CT resulted in higher success rates in histological and molecular successful biopsies compared to CT or MRI. Evaluation of spine biopsies showed more often successful results compared to other locations for both histological and molecular biopsies (p = 0.027 and p = 0.012, respectively). Low Hounsfield units (HUs) and deviation (Dev), taken at CT-guidance, were associated with histological successful biopsies (p = 0.025 and p = 0.023, respectively) and with molecular successful biopsies (p = 0.010 and p = 0.006, respectively). A prediction tool combining low HUs and low Dev resulted in significantly more successful biopsies, histological and molecular (p = 0.023 and p = 0.007, respectively). Based on these results, we concluded that site selection for metastatic tissue biopsies with prior PSMA PET-CT imaging improves the chance of a successful biopsy. Further optimization can be achieved at CT-guidance, by selection of low HU and low Dev lesions. A prediction tool is provided to increase the success rate of bone biopsies in mCRPC patients, which can easily be implemented in daily practice.
Collapse
Affiliation(s)
- Minke Smits
- Department of Medical Oncology, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (K.E.); (S.P.N.); (W.R.G.); (N.M.)
- Correspondence: ; Tel.: +31-24-3618800
| | - Kamer Ekici
- Department of Medical Oncology, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (K.E.); (S.P.N.); (W.R.G.); (N.M.)
| | - Samhita Pamidimarri Naga
- Department of Medical Oncology, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (K.E.); (S.P.N.); (W.R.G.); (N.M.)
| | - Inge M. van Oort
- Department of Urology, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (I.M.v.O.); (M.J.P.S.); (J.A.S.)
| | - Michiel J. P. Sedelaar
- Department of Urology, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (I.M.v.O.); (M.J.P.S.); (J.A.S.)
| | - Jack A. Schalken
- Department of Urology, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (I.M.v.O.); (M.J.P.S.); (J.A.S.)
| | - James Nagarajah
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (J.N.); (T.W.J.S.); (J.J.F.)
- Department of Nuclear Medicine, Technical University, Arcisstraße 21, 80333 Munich, Germany
| | - Tom W. J. Scheenen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (J.N.); (T.W.J.S.); (J.J.F.)
| | - Winald R. Gerritsen
- Department of Medical Oncology, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (K.E.); (S.P.N.); (W.R.G.); (N.M.)
| | - Jurgen J. Fütterer
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (J.N.); (T.W.J.S.); (J.J.F.)
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center Nijmegen, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (K.E.); (S.P.N.); (W.R.G.); (N.M.)
| |
Collapse
|
353
|
Caffo O, Maines F, Kinspergher S, Veccia A, Messina C. To treat or not to treat: is it acceptable to avoid active therapies in advanced prostate cancer today? Expert Rev Anticancer Ther 2020; 21:389-400. [PMID: 33245666 DOI: 10.1080/14737140.2021.1856661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Recently, there has been a paradigm shift in the treatment of advanced prostate cancer (PCa) because the approval of a number of new agents has significantly improved overall survival. However, as PCa is a heterogeneous disease that may be more or less aggressive and patients may be more or less responsive to treatment, it is often debated whether or not it is acceptable to avoid active therapies.Areas covered: This review discusses different settings of advanced PCa.Expert opinion: In metastatic castration-resistant PCa, it is unethical not to use active treatments but the use of both androgen receptor targeting agents (ARTA) in sequence should be avoided in most patients and the use of the available agents for fourth-line treatment or beyond should only be considered for highly selected patients. In metastatic hormone-sensitive PCa, patients with de novo disease should receive one additional agent in combination with androgen deprivation therapy (ADT), whereas patients in relapse should be managed with ADT alone. In non-metastatic castration-resistant prostate cancer (PCa), all patients with a PSA doubling time of ≤6 months should receive one ARTA, whereas the others might wait until there is an acceleration in the kinetics of their PSA levels.
Collapse
Affiliation(s)
- Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| | - Francesca Maines
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| | - Stefania Kinspergher
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| | - Antonello Veccia
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| | - Carlo Messina
- Department of Medical Oncology, Santa Chiara Hospital, Largo Medaglie d'Oro, Trento, Italy
| |
Collapse
|
354
|
Pembrolizumab as Consolidation Strategy in Patients with Multiple Myeloma: Results of the GEM-Pembresid Clinical Trial. Cancers (Basel) 2020; 12:cancers12123615. [PMID: 33287189 PMCID: PMC7761692 DOI: 10.3390/cancers12123615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Multiple myeloma patients with persistent disease after treatment show increased expression of PDL1 in tumor plasma cells and of PD1 in T lymphocytes. This suggests a role of the PD1/PDL1 axis in treatment failure that could potentially be reverted with pembrolizumab, an anti-PD1 monoclonal antibody. The GEM-Pembresid trial enrolled 20 patients with multiple myeloma achieving a suboptimal response to the previous treatment that received intravenous pembrolizumab every 3 weeks with the objective of eradicating the residual disease. Pembrolizumab was acceptably well tolerated in the 17 patients evaluable for safety, but no improvement in the baseline responses was documented. Although no determinants of response could be identified, we detected a lower expression of PD1/PDL1 in a subgroup of patients progressing in the first 4 months after enrollment; furthermore, a reduction in the percentage of NK cells induced by pembrolizumab was observed. Abstract PD1 expression in CD4+ and CD8+ T cells is increased after treatment in multiple myeloma patients with persistent disease. The GEM-Pembresid trial analyzed the efficacy and safety of pembrolizumab as consolidation in patients achieving at least very good partial response but with persistent measurable disease after first- or second-line treatment. Moreover, the characteristics of the immune system were investigated to identify potential biomarkers of response to pembrolizumab. One out of the 17 evaluable patients showed a decrease in the amount of M-protein, although a potential late effect of high-dose melphalan could not be ruled out. Fourteen adverse events were considered related to pembrolizumab, two of which (G3 diarrhea and G2 pneumonitis) prompted treatment discontinuation and all resolving without sequelae. Interestingly, pembrolizumab induced a decrease in the percentage of NK cells at cycle 3, due to the reduction of the circulating and adaptive subsets (0.615 vs. 0.43, p = 0.007; 1.12 vs. 0.86, p = 0.02). In the early progressors, a significantly lower expression of PD1 in CD8+ effector memory T cells (MFI 1327 vs. 926, p = 0.03) was observed. In conclusion, pembrolizumab used as consolidation monotherapy shows an acceptable toxicity profile but did not improve responses in this MM patient population. The trial was registered at clinicaltrials.gov with identifier NCT02636010 and with EUDRACT number 2015-003359-23.
Collapse
|
355
|
Mar N, Kalebasty AR, Uchio EM. Management of Advanced Prostate Cancer in Clinical Practice: Real-World Answers to Challenging Dilemmas. JCO Oncol Pract 2020; 16:783-789. [PMID: 33301698 DOI: 10.1200/op.20.00445] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/12/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022] Open
Abstract
Advanced prostate cancer is a continuum of states distinguished by the presence or absence of metastasis and sensitivity or resistance to androgen deprivation therapy (ADT). Therapeutic options for this disease continue to rapidly evolve, making it a challenge to apply results of clinical trial data to daily patient management. One question relevant to providers is whether molecular biomarkers predictive of response or resistance to therapies are available to guide clinical treatment decisions. Other salient topics include the timing of therapy initiation in patients with biochemical recurrence, treatment approach in low-volume versus high-volume metastatic castrate-sensitive prostate cancer, types of agents available beyond ADT, and timing of their use in men with nonmetastatic castrate-resistant prostate cancer as well as whether sequencing of therapies in metastatic castrate-resistant prostate cancer matters. Additionally, familiarity with emerging treatment strategies is important. This review addresses the gray areas and challenging questions in advanced prostate cancer management that frequently arise in clinical practice.
Collapse
Affiliation(s)
- Nataliya Mar
- Division of Hematology/Oncology, University of California Irvine, Orange, CA
| | | | - Edward M Uchio
- Division of Urology, University of California Irvine, Orange, CA
| |
Collapse
|
356
|
Compérat E, Wasinger G, Oszwald A, Kain R, Cancel-Tassin G, Cussenot O. The Genetic Complexity of Prostate Cancer. Genes (Basel) 2020; 11:E1396. [PMID: 33255593 PMCID: PMC7760266 DOI: 10.3390/genes11121396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 01/09/2023] Open
Abstract
Prostate cancer (PCa) is a major concern in public health, with many genetically distinct subsets. Genomic alterations in PCa are extraordinarily complex, and both germline and somatic mutations are of great importance in the development of this tumor. The aim of this review is to provide an overview of genetic changes that can occur in the development of PCa and their role in potential therapeutic approaches. Various pathways and mechanisms proposed to play major roles in PCa are described in detail to provide an overview of current knowledge.
Collapse
Affiliation(s)
- Eva Compérat
- CeRePP/GRC5 Predictive Onco-Urology, Sorbonne University, 75020 Paris, France; (G.C.-T.); (O.C.)
- Department of Pathology, Hôpital Tenon, Sorbonne University, 75020 Paris, France
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (A.O.); (R.K.)
| | - Gabriel Wasinger
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (A.O.); (R.K.)
| | - André Oszwald
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (A.O.); (R.K.)
| | - Renate Kain
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (A.O.); (R.K.)
| | - Geraldine Cancel-Tassin
- CeRePP/GRC5 Predictive Onco-Urology, Sorbonne University, 75020 Paris, France; (G.C.-T.); (O.C.)
| | - Olivier Cussenot
- CeRePP/GRC5 Predictive Onco-Urology, Sorbonne University, 75020 Paris, France; (G.C.-T.); (O.C.)
- Department of Urology, Hôpital Tenon, Sorbonne University, 75020 Paris, France
| |
Collapse
|
357
|
Vlajnic T, Bubendorf L. Molecular pathology of prostate cancer: a practical approach. Pathology 2020; 53:36-43. [PMID: 33234230 DOI: 10.1016/j.pathol.2020.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
While localised prostate cancer can be cured by local treatment, 'high-risk' prostate cancer often progresses to castration resistant disease and remains incurable with a dismal prognosis. In recent years, technical advances and development of novel methodologies have largely contributed to a better understanding of underlying molecular mechanisms that promote tumour growth and progression. Consecutively, novel therapeutic strategies for treatment of prostate cancer have emerged during the last decade, calling for the identification of predictive biomarkers. The concept of personalised medicine is to tailor treatment according to the specific tumour profile of an individual patient. Moreover, acquired molecular changes during tumour evolution and in response to therapy selection pressure require adapted predictive marker testing at different time points during the disease. In this setting, the pathologist plays a critical role in patient management and treatment selection. In this review, we provide a comprehensive overview of the current knowledge of molecular aspects of prostate cancer and their potential utility in the context of different therapeutic approaches. Furthermore, we discuss methods for molecular marker testing in routine clinical practice, with a focus on castration resistant prostate cancer.
Collapse
Affiliation(s)
- Tatjana Vlajnic
- Institute of Pathology, University Hospital Basel, Basel, Switzerland.
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
358
|
Lin L, Kane N, Kobayashi N, Kono EA, Yamashiro JM, Nickols NG, Reiter RE. High-dose per Fraction Radiotherapy Induces Both Antitumor Immunity and Immunosuppressive Responses in Prostate Tumors. Clin Cancer Res 2020; 27:1505-1515. [PMID: 33219015 DOI: 10.1158/1078-0432.ccr-20-2293] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/13/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE The use of high-dose per fraction radiotherapy delivered as stereotactic body radiotherapy is a standard of care for prostate cancer. It is hypothesized that high-dose radiotherapy may enhance or suppress tumor-reactive immunity. The objective of this study was to assess both antitumor and immunosuppressive effects induced by high-dose radiotherapy in prostate cancer coclinical models, and ultimately, to test whether a combination of radiotherapy with targeted immunotherapy can enhance antitumor immunity. EXPERIMENTAL DESIGN We studied the effects of high-dose per fraction radiotherapy with and without anti-Gr-1 using syngeneic murine allograft prostate cancer models. The dynamic change of immune populations, including tumor-infiltrating lymphocytes (TIL), T regulatory cells (Treg), and myeloid-derived suppressive cells (MDSC), was evaluated using flow cytometry and IHC. RESULTS Coclinical prostate cancer models demonstrated that high-dose per fraction radiotherapy induced a rapid increase of tumor-infiltrating MDSCs and a subsequent rise of CD8 TILs and circulating CD8 T effector memory cells. These radiation-induced CD8 TILs were more functionally potent than those from nonirradiated controls. While systemic depletion of MDSCs by anti-Gr-1 effectively prevented MDSC tumor infiltration, it did not enhance radiotherapy-induced antitumor immunity due to a compensatory expansion of Treg-mediated immune suppression. CONCLUSIONS In allograft prostate cancer models, high-dose radiotherapy induced an early rise of MDSCs, followed by a transient increase of functionally active CD8 TILs. However, systemic depletion of MDSC did not augment the antitumor efficacy of high-dose radiotherapy due to a compensatory Treg response, indicating blocking both MDSCs and Tregs might be necessary to enhance radiotherapy-induced antitumor immunity.
Collapse
Affiliation(s)
- Lin Lin
- Department of Urology, University of California, Los Angeles, Los Angeles, California
| | - Nathanael Kane
- Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, California
| | - Naoko Kobayashi
- Department of Urology, University of California, Los Angeles, Los Angeles, California
| | - Evelyn A Kono
- Department of Urology, University of California, Los Angeles, Los Angeles, California
| | - Joyce M Yamashiro
- Department of Urology, University of California, Los Angeles, Los Angeles, California
| | - Nicholas G Nickols
- Department of Urology, University of California, Los Angeles, Los Angeles, California.,Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, California.,Radiation Oncology Service, VA Greater Los Angeles, Los Angeles, California
| | - Robert E Reiter
- Department of Urology, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
359
|
Norz V, Rausch S. Treatment and resistance mechanisms in castration-resistant prostate cancer: new implications for clinical decision making? Expert Rev Anticancer Ther 2020; 21:149-163. [PMID: 33106066 DOI: 10.1080/14737140.2021.1843430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Introduction: The armamentarium of treatment options in metastatic and non-metastatic CRPC is rapidly evolving. However, the question of how individual treatment decisions should be balanced by available predictive clinical parameters, pharmacogenetic and drug interaction profiles, or compound-associated molecular biomarkers is a major challenge for clinical practice.Areas covered: We discuss treatment and resistance mechanisms in PC with regard to their association to drug efficacy and tolerability. Current efforts of combination treatment and putative predictive biomarkers of available and upcoming compounds are highlighted with regard to their implication on clinical decision-making.Expert opinion: Several treatment approaches are delineated, where identification of resistance mechanisms in CRPC may guide treatment selection. To date, most of these candidate biomarkers will however be found only in a small subset of patients. While current approaches of combination treatment in CRPC are proving synergistic effects on cancer biology, higher complexity with regard to biomarker analysis and interaction profiles of the respective compounds may be expected. Among other aspects of personalized treatment, consideration of drug-drug interaction and pharmacogenetics is an underrepresented issue. However, the non-metastatic castration resistant prostate cancer situation may be an example for treatment selection based on drug interaction profiles in the future.
Collapse
Affiliation(s)
- Valentina Norz
- Department of Urology, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - Steffen Rausch
- Department of Urology, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| |
Collapse
|
360
|
Wang Z, Zhao X, Gao C, Gong J, Wang X, Gao J, Li Z, Wang J, Yang B, Wang L, Zhang B, Zhou Y, Wang D, Li X, Bai Y, Li J, Shen L. Plasma-based microsatellite instability detection strategy to guide immune checkpoint blockade treatment. J Immunother Cancer 2020; 8:jitc-2020-001297. [PMID: 33172882 PMCID: PMC7656957 DOI: 10.1136/jitc-2020-001297] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2020] [Indexed: 01/10/2023] Open
Abstract
Background Microsatellite instability (MSI) represents the first pan-cancer biomarker approved to guide immune checkpoint blockade (ICB) treatment. However its widespread testing, especially outside of gastrointestinal cancer, is hampered by tissue availability. Methods An algorithm for detecting MSI from peripheral blood was established and validated using clinical plasma samples. Its value for predicting ICB efficacy was evaluated among 60 patients with advanced gastrointestinal cancer. The landscape of MSI in blood was also explored among 5138 advanced solid tumors. Results The algorithm included 100 microsatellite markers with high capture efficiency, sensitivity, and specificity. In comparison with orthogonal tissue PCR results, the method displayed a sensitivity of 82.5% (33/40) and a specificity of 96.2% (201/209), for an overall accuracy of 94.0% (234/249). When the clinical validation cohort was dichotomized by pretreatment blood MSI (bMSI), bMSI-high (bMSI-H) predicted both improved progression-free survival and overall survival than the blood microsatellite stable (bMSS) patients (HRs: 0.431 and 0.489, p=0.005 and 0.034, respectively). Four patients with bMSS were identified to have high blood tumor mutational burden (bTMB-H) and trended towards a better survival than the bMSS-bTMB-low (bTMB-L) subset (HR 0.026, 95% CI 0 to 2.635, p=0.011). These four patients with bMSS-bTMB-H plus the bMSI-H group collectively displayed significantly improved survival over the bMSS-bTMB-L patients (HR 0.317, 95% CI 0.157 to 0.640, p<0.001). Pan-cancer prevalence of bMSI-H was largely consistent with that shown for tissue except for much lower rates in endometrial and gastrointestinal cancers, and a remarkably higher prevalence in prostate cancer relative to other cancer types. Conclusions We have developed a reliable and robust next generation sequencing-based bMSI detection strategy which, in combination with a panel enabling concurrent profiling of bTMB from a single blood draw, may better inform ICB treatment.
Collapse
Affiliation(s)
- Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | | | - Chan Gao
- Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xicheng Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing Gao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhongwu Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Wang
- Research & Development, 3D Medicines Inc, Shanghai, China
| | - Bo Yang
- Research & Development, 3D Medicines Inc, Shanghai, China
| | - Lei Wang
- Research & Development, 3D Medicines Inc, Shanghai, China
| | - Bei Zhang
- Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Yifan Zhou
- Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Dalei Wang
- Clinical Laboratory, 3D Medicines Inc, Shanghai, China
| | - Xiaofang Li
- Research & Development, 3D Medicines Inc, Shanghai, China
| | - Yuezong Bai
- Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
361
|
Halbert B, Einstein DJ. Hot or Not: Tumor Mutational Burden (TMB) as a Biomarker of Immunotherapy Response in Genitourinary Cancers. Urology 2020; 147:119-126. [PMID: 33137348 DOI: 10.1016/j.urology.2020.10.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/31/2022]
Abstract
Pembrolizumab was recently approved for treatment of cancers with high tumor mutational burden (TMB). We conduct a focused literature review of TMB as a predictive biomarker. TMB quantifies the sum of nonsynonymous coding mutations (typically single nucleotide substitutions and short insertion-deletions) per megabase of sequenced DNA. As a proxy for expression of immunogenic neoantigens, TMB may be an effective predictive biomarker for response to immune checkpoint inhibitors. However, like other biomarkers in this setting, TMB has many limitations; the effect of this FDA approval in the current management of genitourinary cancers is likely limited to select situations.
Collapse
Affiliation(s)
- Brian Halbert
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA
| | - David J Einstein
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA.
| |
Collapse
|
362
|
Burdak-Rothkamm S, Mansour WY, Rothkamm K. DNA Damage Repair Deficiency in Prostate Cancer. Trends Cancer 2020; 6:974-984. [DOI: 10.1016/j.trecan.2020.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022]
|
363
|
Simnica D, Smits M, Willscher E, Fanchi LF, Kloots ISH, van Oort I, Gerritsen W, Mehra N, Binder M. Responsiveness to Immune Checkpoint Inhibitors Is Associated With a Peripheral Blood T-Cell Signature in Metastatic Castration-Resistant Prostate Cancer. JCO Precis Oncol 2020; 4:1374-1385. [PMID: 35050788 DOI: 10.1200/po.20.00209] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Although most patients with microsatellite instable (MSI) metastatic castration-resistant prostate cancer (mCRPC) respond to immune checkpoint blockade (ICB), only a small subset of patients with microsatellite stable (MSS) tumors have similar benefit. Biomarkers defining ICB-susceptible subsets of patients with MSS mCRPC are urgently needed. METHODS Using next-generation T-cell repertoire sequencing, we explored immune signatures in 54 patients with MSS and MSI mCRPC who were treated with or without ICB. We defined subset-specific immune metrics as well as T-cell clusters and correlated the signatures with treatment benefit. RESULTS Consistent overlaps between tumor and peripheral T-cell repertoires suggested that blood was an informative material to identify relevant T-cell signatures. We found considerably higher blood T-cell richness and diversity and more shared T-cell clusters with low generation probability (pGen) in MSI versus MSS mCRPC, potentially reflecting more complex T-cell responses because of a greater neoepitope load in the MSI subset. Interestingly, patients with MSS mCRPC with shared low pGen T-cell clusters showed significantly better outcomes with ICB, but not with other treatments, compared with patients without such clusters. Blood clearance of T-cell clusters on ICB treatment initiation seemed to be compatible with T-cell migration to the primary tumor or metastatic sites during the process of clonal replacement as described for other tumors receiving ICB. CONCLUSION The MSI mCRPC subset shows a distinct T-cell signature that can be detected in blood. This signature points to immune parameters that could help identify a subset of patients with MSS mCRPC who may have an increased likelihood of responding to ICB or to combination approaches including ICB.
Collapse
Affiliation(s)
- Donjete Simnica
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Minke Smits
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Edith Willscher
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | | | - Iris S H Kloots
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Inge van Oort
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Winald Gerritsen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
364
|
McGregor BA, Wei XX. CDK12 Loss in Prostate Cancer Imparts Poor Prognosis and Limited Susceptibility to Immune Checkpoint Inhibition. JCO Precis Oncol 2020; 4:367-369. [DOI: 10.1200/po.20.00080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
365
|
Mateo J, McKay R, Abida W, Aggarwal R, Alumkal J, Alva A, Feng F, Gao X, Graff J, Hussain M, Karzai F, Montgomery B, Oh W, Patel V, Rathkopf D, Rettig M, Schultz N, Smith M, Solit D, Sternberg C, Van Allen E, VanderWeele D, Vinson J, Soule HR, Chinnaiyan A, Small E, Simons JW, Dahut W, Miyahira AK, Beltran H. Accelerating precision medicine in metastatic prostate cancer. NATURE CANCER 2020; 1:1041-1053. [PMID: 34258585 PMCID: PMC8274325 DOI: 10.1038/s43018-020-00141-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
Despite advances in prostate cancer screening and treatment, available therapy options, particularly in later stages of the disease, remain limited and the treatment-resistant setting represents a serious unmet medical need. Moreover, disease heterogeneity and disparities in patient access to medical advances result in significant variability in outcomes across patients. Disease classification based on genomic sequencing is a promising approach to identify patients whose tumors exhibit actionable targets and make more informed treatment decisions. Here we discuss how we can accelerate precision oncology to inform broader genomically-driven clinical decisions for men with advanced prostate cancer, drug development and ultimately contribute to new treatment paradigms.
Collapse
Affiliation(s)
- Joaquin Mateo
- Vall d'Hebron Institute of Oncology and Vall d'Hebron University Hospital, Barcelona, Spain
| | - Rana McKay
- University of California at San Diego, San Diego, CA, USA
| | - Wassim Abida
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rahul Aggarwal
- University of California at San Francisco, San Francisco, CA, USA
| | | | - Ajjai Alva
- University of Michigan, Ann Arbor, MI, USA
| | - Felix Feng
- University of California at San Francisco, San Francisco, CA, USA
| | - Xin Gao
- Massachusetts General Hospital, Boston, MA, USA
| | - Julie Graff
- Oregon Health & Science University, VA Portland Health Care System, Portland, OR, USA
| | - Maha Hussain
- Lurie Comprehensive Cancer Center at Northwestern University, Chicago, IL, USA
| | | | | | | | | | - Dana Rathkopf
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew Rettig
- University of California at Los Angeles, VA Greater Los Angeles, Los Angeles, CA, USA
| | | | | | - David Solit
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - David VanderWeele
- Lurie Comprehensive Cancer Center at Northwestern University, Chicago, IL, USA
| | - Jake Vinson
- Prostate Cancer Clinical Trials Consortium, New York, NY, USA
| | | | | | - Eric Small
- University of California at San Francisco, San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
366
|
Recommandations françaises du Comité de cancérologie de l’AFU – actualisation 2020–2022 : cancer de la prostate. Prog Urol 2020; 30:S136-S251. [DOI: 10.1016/s1166-7087(20)30752-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
367
|
Powers E, Karachaliou GS, Kao C, Harrison MR, Hoimes CJ, George DJ, Armstrong AJ, Zhang T. Novel therapies are changing treatment paradigms in metastatic prostate cancer. J Hematol Oncol 2020; 13:144. [PMID: 33115529 PMCID: PMC7594418 DOI: 10.1186/s13045-020-00978-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/11/2020] [Indexed: 12/11/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains a terminal diagnosis with an aggressive disease course despite currently approved therapeutics. The recent successful development of poly ADP-ribose polymerase (PARP) inhibitors for patients with mCRPC and mutations in DNA damage repair genes has added to the treatment armamentarium and improved personalized treatments for prostate cancer. Other promising therapeutic agents currently in clinical development include the radiotherapeutic 177-lutetium-prostate-specific membrane antigen (PSMA)-617 targeting PSMA-expressing prostate cancer and combinations of immunotherapy with currently effective treatment options for prostate cancer. Herein, we have highlighted the progress in systemic treatments for mCRPC and the promising agents currently in ongoing clinical trials.
Collapse
Affiliation(s)
- Eric Powers
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Georgia Sofia Karachaliou
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Chester Kao
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Michael R Harrison
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Christopher J Hoimes
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Daniel J George
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Andrew J Armstrong
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA.,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA. .,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA.
| |
Collapse
|
368
|
Lang D, Wahl G, Poier N, Graf S, Kiesl D, Lamprecht B, Gabriel M. Impact of PET/CT for Assessing Response to Immunotherapy-A Clinical Perspective. J Clin Med 2020; 9:jcm9113483. [PMID: 33126715 PMCID: PMC7694130 DOI: 10.3390/jcm9113483] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/18/2020] [Accepted: 10/22/2020] [Indexed: 12/25/2022] Open
Abstract
Cancer immunotherapy using immune-checkpoint inhibitors (ICI) has revolutionized the therapeutic landscape of various malignancies like non-small-cell lung cancer or melanoma. Pre-therapy response prediction and assessment during ICI treatment is challenging due to the lack of reliable biomarkers and the possibility of atypical radiological response patterns. Positron emission tomography/computed tomography (PET/CT) enables the visualization and quantification of metabolic lesion activity additional to conventional CT imaging. Various biomarkers derived from PET/CT have been reported as predictors for response to ICI and may aid to overcome the challenges clinicians currently face in the management of ICI-treated patients. In this narrative review, experts in nuclear medicine, thoracic oncology, dermatooncology, hemato- and internal oncology, urological and head/neck tumors performed literature reviews in their respective field and a joint discussion on the use of PET/CT in the context of ICI treatment. The aims were to give a clinical overview on present standards and evidence, to identify current challenges and fields of research and to enable an outlook to future developments and their possible implications. Multiple promising studies concerning ICI response assessment or prediction using biomarkers derived from PET/CT alone or as composite biomarkers have been identified for various malignancies and disease stages. Of interest, additional major incentives in the field may evolve from novel tracers specifically targeting immune-checkpoint molecules which could allow not only response assessment and prognosis, but also visualization of histological tumor cell properties like programmed death-ligand (PD-L1) expression in vivo. Despite the broad range of existing literature on PET/CT-derived biomarkers in ICI therapy, implications for daily clinical practice remain elusive. High-quality prospective data are urgently warranted to determine whether patients benefit from the application of PET/CT in terms of prognosis. At the moment, the lack of such evidence as well as the absence of standardized imaging methods and biomarkers still precludes PET/CT imaging to be included in the relevant clinical practice guidelines.
Collapse
Affiliation(s)
- David Lang
- Department of Pulmonology Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria; (D.L.); (B.L.)
| | - Gerald Wahl
- Department of Dermatology and Venerology, Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria;
| | - Nikolaus Poier
- Department of Otorhinolaryngology, Head and Neck Surgery, Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria;
| | - Sebastian Graf
- Department of Urology and Andrology, Johannes Kepler University Hospital Linz Krankenhausstrasse 9, 4020 Linz, Austria;
| | - David Kiesl
- University Clinic of Hematology and Internal Oncology Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria;
| | - Bernd Lamprecht
- Department of Pulmonology Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria; (D.L.); (B.L.)
| | - Michael Gabriel
- Institute of Nuclear Medicine and Endocrinology, Johannes Kepler University Hospital Linz, Krankenhausstrasse 9, 4020 Linz, Austria
- Correspondence: ; Tel.: +43-5-7680-83-6166; Fax: +43-5-7680-83-6165
| |
Collapse
|
369
|
Tsaur I, Brandt MP, Juengel E, Manceau C, Ploussard G. Immunotherapy in prostate cancer: new horizon of hurdles and hopes. World J Urol 2020; 39:1387-1403. [PMID: 33106940 PMCID: PMC8514362 DOI: 10.1007/s00345-020-03497-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Prostate cancer (PCa) is the most common malignancy in men and the cause for the second most common cancer-related death in the western world. Despite ongoing development of novel approaches such as second generation androgen receptor targeted therapies, metastatic disease is still fatal. In PCa, immunotherapy (IT) has not reached a therapeutic breakthrough as compared to several other solid tumors yet. We aimed at highlighting the underlying cellular mechanisms crucial for IT in PCa and giving an update of the most essential past and ongoing clinical trials in the field. Methods We searched for relevant publications on molecular and cellular mechanisms involved in the PCa tumor microenvironment and response to IT as well as completed and ongoing IT studies and screened appropriate abstracts of international congresses. Results Tumor progression and patient outcomes depend on complex cellular and molecular interactions of the tumor with the host immune system, driven rather dormant in case of PCa. Sipuleucel-T and pembrolizumab are the only registered immune-oncology drugs to treat this malignancy. A plethora of studies assess combination of immunotherapy with other agents or treatment modalities like radiation therapy which might increase its antineoplastic activity. No robust and clinically relevant prognostic or predictive biomarkers have been established yet. Conclusion Despite immunosuppressive functional status of PCa microenvironment, current evidence, based on cellular and molecular conditions, encourages further research in this field.
Collapse
Affiliation(s)
- Igor Tsaur
- Department of Urology and Pediatric Urology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Maximilian P Brandt
- Department of Urology and Pediatric Urology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Eva Juengel
- Department of Urology and Pediatric Urology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Cécile Manceau
- Department of Urology, CHU-Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Guillaume Ploussard
- Department of Urology, CHU-Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France.,Department of Urology, La Croix du Sud Hospital, Toulouse, France
| |
Collapse
|
370
|
Sardana R, Mishra SK, Williamson SR, Mohanty A, Mohanty SK. Immune checkpoints and their inhibitors: Reappraisal of a novel diagnostic and therapeutic dimension in the urologic malignancies. Semin Oncol 2020; 47:367-379. [PMID: 33160642 DOI: 10.1053/j.seminoncol.2020.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
Abstract
Advances in molecular immunology have unveiled some of the complexity of the mechanisms regulating cellular immune responses and led to the successful targeting of immune checkpoints in attempts to enhance antitumor T cell responses. Surgery, chemotherapy, and radiation therapy have been the mainstay of treatment in urologic malignancies. Immune checkpoint molecules such as cytotoxic T-lymphocyte associated protein-4, programmed cell death protein-1, and programmed death-ligand 1 have been shown to play central roles in evading cancer immunity. Thus these molecules have been targeted by inhibitors for the management of cancers forming the basis of immunotherapy. Immunotherapy is now among the first line therapeutic options for metastatic renal cell carcinomas. In advanced bladder cancer, immunotherapy is the standard of care in the second line and the first line for cisplatin ineligible patients. There continues to be ongoing research to identify the role if any of immunotherapy in testicular, prostatic, and penile cancers. The ideal biomarker for response to immunotherapy is still elusive. Although programmed death-ligand 1 immunohistochemical testing has been widely used across the globe as a biomarker for immunotherapy, companion diagnostic tests have inherent issues with testing and reporting and cannot have universal applicability. Additional biomarkers including, tumor mutational burden, deficient mismatch repair, high microsatellite instability, and immune gene expression profiling are being evaluated in various clinical trials. This review appraises the data of immunotherapy in the management of urologic malignancies.
Collapse
Affiliation(s)
- Rohan Sardana
- Department of Hematopathology, Tata Memorial Hospital, Mumbai, India
| | - Sourav K Mishra
- Department of Medical Oncology, Advanced Medical Research Institute, Bhubaneswar, India
| | - Sean R Williamson
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, MI, USA
| | - Abhishek Mohanty
- Principal Research Officer Head of Research, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India.
| |
Collapse
|
371
|
Ng CF, Chiu PKF, Yee CH, Lau BSY, Leung SCH, Teoh JYC. Effect of androgen deprivation therapy on cardiovascular function in Chinese patients with advanced prostate cancer: a prospective cohort study. Sci Rep 2020; 10:18060. [PMID: 33093594 PMCID: PMC7582924 DOI: 10.1038/s41598-020-75139-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/12/2020] [Indexed: 11/09/2022] Open
Abstract
Androgen deprivation therapy (ADT) is the standard treatment for advanced prostate cancer, but its effect on cardiovascular and metabolic function in Asian patients is still inconclusive. We prospectively assess the effects of ADT on 36 patients with advanced prostate cancer, with reference to another 24 prostate cancer patients not requiring ADT, for 2 years. Patients’ anthropometric, metabolic and vascular parameters were assessed every six-monthly. The baseline parameters of the two groups were comparable. There was a significant negative effect of the usage of ADT on the changes in BMI (p = 0.020), waist to hip ratio (p = 0.005), body fat percentage (p = 0.012), and high-density-lipoprotein (p = 0.012). ADT-patients were 4.9 times more likely to have metabolic syndrome at 24 months. (CI 0.889–27.193, p = 0.068). The Framingham risk score (p = 0.018) and pulse-wave-velocity (p = 0.024) for ADT-group were also significantly higher than controls, which signified increase in cardiovascular risk. Although there was no statistically significant difference in ischemic cardiovascular events between two groups, a trend for more events in ADT-group was observed. Therefore, Asian patients have increased cardiovascular and metabolic risks after being treated with ADT for two years. Appropriate counselling and monitoring of associated complications with ADT are essential.
Collapse
Affiliation(s)
- Chi-Fai Ng
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Peter K F Chiu
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi-Hang Yee
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Becky S Y Lau
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Steven C H Leung
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jeremy Y C Teoh
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
372
|
Report From the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers. I. Molecular Biomarkers in Prostate Cancer. Am J Surg Pathol 2020; 44:e15-e29. [PMID: 32044806 DOI: 10.1097/pas.0000000000001450] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The combined clinical and molecular heterogeneity of prostate cancer necessitates the use of prognostic, predictive, and diagnostic biomarkers to assist the clinician with treatment selection. The pathologist plays a critical role in guiding molecular biomarker testing in prostate cancer and requires a thorough knowledge of the current testing options. In the setting of clinically localized prostate cancer, prognostic biomarkers such as Ki-67 labeling, PTEN loss or mRNA-based genomic signatures can be useful to help determine whether definitive therapy is required. In the setting of advanced disease, predictive biomarkers, such as the presence of DNA repair deficiency mediated by BRCA2 loss or mismatch repair gene defects, may suggest the utility of poly-ADP ribosylase inhibition or immune checkpoint blockade. Finally, androgen receptor-related biomarkers or diagnostic biomarkers indicating the presence of small cell neuroendocrine prostate cancer may help guide the use of androgen receptor signaling inhibitors and chemotherapy. In this review, we examine the current evidence for several prognostic, predictive and diagnostic tissue-based molecular biomarkers in prostate cancer management. For each assay, we summarize a recent survey of the International Society of Urology Pathology (ISUP) members on current testing practices and include recommendations for testing that emerged from the ISUP Working Group on Molecular Pathology of Prostate Cancer and the 2019 Consultation Conference on Molecular Pathology of Urogenital Cancers.
Collapse
|
373
|
Sharma P, Pachynski RK, Narayan V, Fléchon A, Gravis G, Galsky MD, Mahammedi H, Patnaik A, Subudhi SK, Ciprotti M, Simsek B, Saci A, Hu Y, Han GC, Fizazi K. Nivolumab Plus Ipilimumab for Metastatic Castration-Resistant Prostate Cancer: Preliminary Analysis of Patients in the CheckMate 650 Trial. Cancer Cell 2020; 38:489-499.e3. [PMID: 32916128 DOI: 10.1016/j.ccell.2020.08.007] [Citation(s) in RCA: 242] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/14/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is immunologically "cold" and predominantly resistant to immune checkpoint therapy due to few tumor-infiltrating T cells. Ipilimumab (anti-CTLA-4) or anti-PD-1/PD-L1 monotherapy failed to show a significant benefit. Although the PD-1/PD-L1 pathway is minimally expressed in prostate tumors, we previously demonstrated that PD-1/PD-L1 expression increases as a compensatory inhibitory pathway in parallel with an ipilimumab-induced increase in tumor-infiltrating T cells. Here, we report the largest trial to date in mCRPC with anti-CTLA-4 plus anti-PD-1 (nivolumab 1 mg/kg plus ipilimumab 3 mg/kg; CheckMate 650, NCT02985957). With median follow-ups of 11.9 and 13.5 months in cohorts 1 (pre-chemotherapy; n = 45) and 2 (post-chemotherapy; n = 45), objective response rate was 25% and 10%, and median overall survival was 19.0 and 15.2 months, respectively. Four patients, two in each cohort, had complete responses. Exploratory studies identify potential biomarkers of response. Grade 3-4 treatment-related adverse events have occurred in ∼42%-53% of patients, with four treatment-related deaths. Therefore, dose/schedule modifications have been implemented.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Unit 1374, 1155 Pressler Street, Houston, TX 77030, USA.
| | - Russell K Pachynski
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vivek Narayan
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aude Fléchon
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes Aix-Marseille Université, 13009 Marseille, France
| | - Matthew D Galsky
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hakim Mahammedi
- Department of Medical Oncology, Centre Jean Perrin, 63011 Clermont-Ferrand, France
| | - Akash Patnaik
- Department of Medical Oncology, The University of Chicago Medicine, Chicago, IL 60637, USA
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, University of Texas, Unit 1374, 1155 Pressler Street, Houston, TX 77030, USA
| | | | | | - Abdel Saci
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | - Yanhua Hu
- Bristol Myers Squibb, Princeton, NJ 08540, USA
| | | | - Karim Fizazi
- Department of Cancer Medicine, Gustave Roussy, University of Paris Saclay, 94800 Villejuif, France
| |
Collapse
|
374
|
Ye S, Wang H, He K, Peng M, Wang Y, Li Y, Jiang S, Li J, Yi L, Cui R. Clinical Characterization of Mismatch Repair Gene-Deficient Metastatic Castration-Resistant Prostate Cancer. Front Oncol 2020; 10:533282. [PMID: 33117677 PMCID: PMC7576180 DOI: 10.3389/fonc.2020.533282] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/24/2020] [Indexed: 11/13/2022] Open
Abstract
Mismatch repair-deficient (dMMR) prostate cancer is rare and has not been well studied. We aimed to evaluate the clinical characterization of dMMR metastatic castration-resistant prostate cancer (mCRPC) patients. The MMR genes include MLH1, MLH3, MSH2, MSH6, PMS1, PMS2, and EPCAM, and were analyzed by targeted sequencing of plasma cell-free DNA samples. A total of 109 mCRPC patients were identified, including 50 patients with MMR alterations (pathogenic alterations, n = 7; alterations of unknown significance, n = 43) and 59 patients with wild-type MMR. For the seven patients with pathogenic MMR alterations, the median age at diagnosis was 63.5 years, and 42.9% had a Gleason score ≥8. The median time from androgen deprivation therapy (ADT) initiation to CRPC was 24 months. Compared with the wild-type MMR subgroup, patients with MMR alterations, pathogenic MMR alterations, or MMR alterations of unknown significance showed higher rates of hotspot missense mutations or copy number amplifications in the AR gene (24/50 vs. 10/59, P = 7.8 × 10–4; 7/7 vs. 10/59, P = 2.5 × 10–5; 17/43 vs. 10/59, P = 0.013). The presence of any MMR alterations was associated with an inferior response to abiraterone [median progression-free survival (PFS): 5.0 vs. 10.9 months, P = 0.022]. Shorter PFS times were observed in both the pathogenic MMR alteration subgroup (median PFS: 5 months) and the MMR alterations of unknown significance subgroup (median PFS: 5.3 months), compared with the PFS of those with wild-type MMR genes (median PFS: 10.9 months, P = 0.052). There was no statistically significant difference in response to docetaxel chemotherapy between the MMR alterations of unknown significance and the wild-type MMR subgroups (median PFS: 8.2 vs. 8.1 months, P = 0.23). Our results demonstrate that dMMR mCRPC patients have an equivalent response to standard ADT and taxane-based chemotherapy treatments compared with wild-type MMR patients. Patients with both pathogenic and unknown significance alterations of MMR genes had poorer responses to abiraterone therapy.
Collapse
Affiliation(s)
- Senlin Ye
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haohui Wang
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Kancheng He
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Mou Peng
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yuanwei Li
- Department of Urology, Hunan Provincial People's Hospital, Changsha, China
| | - Shusuan Jiang
- Department of Urology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jin Li
- Department of Urology, Central Hospital of Xiangtan, Changsha, China
| | - Lu Yi
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Rongrong Cui
- Department of Metabolism and Endocrinology, Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
375
|
Shore ND, Drake CG, Lin DW, Ryan CJ, Stratton KL, Dunshee C, Karsh LI, Kaul S, Kernen K, Pieczonka C, Sieber P, Stewart C, Williams M, Concepcion RS. Optimizing the management of castration-resistant prostate cancer patients: A practical guide for clinicians. Prostate 2020; 80:1159-1176. [PMID: 32779781 DOI: 10.1002/pros.24053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/27/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Advanced prostate cancer (PC) patients, especially those with metastatic prostate cancer (mPC), often require complex management pathways. Despite the publication of clinical practice guidelines by leading urological and oncological organizations that provide a substantial and comprehensive framework, there are numerous clinical scenarios that are not always addressed, especially as new treatments become available, new imaging modalities are developed, and advances in genetic testing continue. METHODS A 14-member expert review panel comprised of urologists and medical oncologists were chosen to provide guidance on addressing specific topics and issues regarding metastatic castration-resistant prostate cancer (mCRPC) patients. Panel members were chosen based upon their experience and expertise in the management of PC patients. Four academic members (two urologists and two medical oncologists) of the panel served as group leaders; the remaining eight panel members were from Large Urology Group Practice Association (LUGPA) practices with proven experience in leading their advanced PC clinics. The panel members were assigned to four separate working groups, each assigned a specific mCRPC topic to review and discuss with the entire panel. RESULTS This article describes the practical recommendations of an expert panel on the management of mCRPC patients. The target reading audience for this publication is all providers (urologists, medical oncologists, radiation oncologists, or advanced practice providers) who evaluate and manage advanced PC patients, regardless of their practice setting. CONCLUSION The panel has provided recommendations for managing mCRPC with regard to specific issues: (a) biomarker monitoring and the role of genetic and molecular testing; (b) rationale, current strategies, and optimal sequencing of the various approved therapies, including hormonal therapy, cytotoxic chemotherapy, radiopharmaceuticals and immunotherapy; (c) adverse event management and monitoring; and (d) imaging advanced PC patients. These recommendations seek to complement national guidelines, not replace them, and a discussion of where the panel agreed or disagreed with national guidelines is included.
Collapse
Affiliation(s)
- Neal D Shore
- Carolina Urologic Research Center, Atlantic Urology Clinics, Myrtle Beach, South Carolina
| | | | | | - Charles J Ryan
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Kelly L Stratton
- Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Curtis Dunshee
- Urological Associates of Southern Arizona, Tucson, Arizona
| | | | - Sanjeev Kaul
- Michigan Healthcare Professionals, Troy, Michigan
| | - Ken Kernen
- Michigan Institute of Urology, Detroit, Michigan
| | | | - Paul Sieber
- Keystone Urology Specialists, Lancaster, Pennsylvania
| | | | | | - Raoul S Concepcion
- Integra Connect, West Palm Beach, Florida
- Department of Urology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
376
|
Johnson E, Nussenzveig R, Agarwal N, Swami U. Germline variants and response to systemic therapy in advanced prostate cancer. Pharmacogenomics 2020; 21:75-81. [PMID: 31849283 DOI: 10.2217/pgs-2019-0125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Our current understanding of prostate cancer pharmacogenomics is growing at a rapid pace. Apart from evaluating relevant biomarkers and genomic alterations in tumor tissues, an increasing focus is being placed on decoding the impact of germline alterations on prostate cancer and its treatment. Herein we summarize various germline variants that have shown to associate with response to systemic therapy in men with advanced prostate cancer. Covered biomarkers include HSD3B1, SLCO2B1, SULT1E1, TRMT11, CYP17A1, CYP1B1, genes involved in homologous recombination and DNA mismatch repair.
Collapse
Affiliation(s)
- Eric Johnson
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Roberto Nussenzveig
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
377
|
Patel D, McKay R, Parsons JK. Immunotherapy for Localized Prostate Cancer: The Next Frontier? Urol Clin North Am 2020; 47:443-456. [PMID: 33008495 DOI: 10.1016/j.ucl.2020.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer vaccines, cytokines, and checkpoint inhibitors are immunotherapeutic agents that act within the cancer immunity cycle. Prostate cancer has provided unique opportunities for, and challenges to, immunotherapy drug development, including low tumor mutational burdens, limited expression of PD-L1, and minimal T-cell intratumoral infiltrates. Nevertheless, efforts are ongoing to help prime prostate tumors by turning a "cold" prostate cancer "hot" and thus rendering them more susceptible to immunotherapy. Combination treatments, use of molecular biomarkers, and use of new immunotherapeutic agents provide opportunities to enhance the immune response to prostate tumors.
Collapse
Affiliation(s)
- Devin Patel
- Department of Urology, University of California San Diego, 9400 Campus Point Drive, MC7987, La Jolla, CA 92093, USA
| | - Rana McKay
- Division of Hematology-Oncology, Department of Internal Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - J Kellogg Parsons
- Department of Urology, University of California San Diego, 9400 Campus Point Drive, MC7987, La Jolla, CA 92093, USA.
| |
Collapse
|
378
|
Rizzo A, Mollica V, Cimadamore A, Santoni M, Scarpelli M, Giunchi F, Cheng L, Lopez-Beltran A, Fiorentino M, Montironi R, Massari F. Is There a Role for Immunotherapy in Prostate Cancer? Cells 2020; 9:E2051. [PMID: 32911806 PMCID: PMC7564598 DOI: 10.3390/cells9092051] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 02/06/2023] Open
Abstract
In the last decade, immunotherapy has revolutionized the treatment landscape of several hematological and solid malignancies, reporting unprecedented response rates. Unfortunately, this is not the case for metastatic castration-resistant prostate cancer (mCRPC), as several phase I and II trials assessing programmed death receptor 1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors have shown limited benefits. Moreover, despite sipuleucel-T representing the only cancer vaccine approved by the Food and Drug Administration (FDA) for mCRPC following the results of the IMPACT trial, the use of this agent is relatively limited in everyday clinical practice. The identification of specific histological and molecular biomarkers that could predict response to immunotherapy represents one of the current challenges, with an aim to detect subgroups of mCRPC patients who may benefit from immune checkpoint monoclonal antibodies as monotherapy or in combination with other anticancer agents. Several unanswered questions remain, including the following: is there-or will there ever be-a role for immunotherapy in prostate cancer? In this review, we aim at underlining the failures and promises of immunotherapy in prostate cancer, summarizing the current state of art regarding cancer vaccines and immune checkpoint monoclonal antibodies, and discussing future research directions in this immunologically "cold" malignancy.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni, 40138 Bologna, Italy; (A.R.); (V.M.)
| | - Veronica Mollica
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni, 40138 Bologna, Italy; (A.R.); (V.M.)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62012 Macerata, Italy;
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Francesca Giunchi
- Department of Pathology, Ospedale Maggiore and University of Bologna, 40138 Bologna, Italy; (F.G.); (M.F.)
| | - Liang Cheng
- Laboratory Medicine and Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Michelangelo Fiorentino
- Department of Pathology, Ospedale Maggiore and University of Bologna, 40138 Bologna, Italy; (F.G.); (M.F.)
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Francesco Massari
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni, 40138 Bologna, Italy; (A.R.); (V.M.)
| |
Collapse
|
379
|
Handa S, Hans B, Goel S, Bashorun HO, Dovey Z, Tewari A. Immunotherapy in prostate cancer: current state and future perspectives. Ther Adv Urol 2020; 12:1756287220951404. [PMID: 32952615 PMCID: PMC7476347 DOI: 10.1177/1756287220951404] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/25/2020] [Indexed: 02/06/2023] Open
Abstract
Metastatic castrate resistant prostate cancer (PCa) remains an incurable entity. In the era of immunotherapy, the complex PCa microenvironment poses a unique challenge to the successful application of this class of agents. However, in the last decade, a tremendous effort has been made to explore this field of therapeutics. In this review, the physiology of the cancer immunity cycle is highlighted in the context of the prostate tumor microenvironment, and the current evidence for use of various classes of immunotherapy agents including vaccines (dendritic cell based, viral vector based and DNA/mRNA based), immune checkpoint inhibitors, Chimeric antigen receptor T cell therapy, antibody-mediated radioimmunotherapy, antibody drug conjugates, and bispecific antibodies, is consolidated. Finally, the future directions for combinatorial approaches to combat PCa are discussed.
Collapse
Affiliation(s)
- Shivani Handa
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West Hospital, New York, NY, 10019, USA
| | - Bandhul Hans
- Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Shokhi Goel
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Hafis O Bashorun
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Zach Dovey
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Ashutosh Tewari
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
380
|
Barata P, Agarwal N, Nussenzveig R, Gerendash B, Jaeger E, Hatton W, Ledet E, Lewis B, Layton J, Babiker H, Bryce A, Garje R, Stein C, Kiedrowski L, Saylor P, Sartor O. Clinical activity of pembrolizumab in metastatic prostate cancer with microsatellite instability high (MSI-H) detected by circulating tumor DNA. J Immunother Cancer 2020; 8:jitc-2020-001065. [PMID: 32788235 PMCID: PMC7422632 DOI: 10.1136/jitc-2020-001065] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2020] [Indexed: 12/16/2022] Open
Abstract
To report a multi-institutional case series of patients with advanced microsatellite instability high (MSI-H) prostate adenocarcinoma identified with clinical cell-free DNA (cfDNA) next-generation sequencing (NGS) testing and treated with immune checkpoint inhibitors. Retrospective analysis of patients with metastatic castration-resistant prostate cancer (mCRPC) and MSI-H tumor detected by a commercially available cfDNA NGS assay Guardant360 (G360, Guardant Health) at eight different Academic Institutions in the USA, from September 2018 to April 2020. From a total of 14 MSI-H metastatic prostate cancer patients at participating centers, nine patients with mCRPC with 56% bone, 33% nodal, 11% liver and 11% soft-tissue metastases and a median PSA of 29.3 ng/dL, were treated with pembrolizumab after 2 lines of therapy for CRPC. The estimated median time on pembrolizumab was 9.9 (95% CI 1.0 to 18.8) months. Four patients (44%) achieved PSA50 after a median of 4 (3–12) weeks after treatment initiation including three patients with >99% PSA decline. Among the patients evaluable for radiographic response (n=5), the response rate was 60% with one complete response and two partial responses. Best response was observed after a median of 3.3 (1.4–7.6) months. At time of cut-off, four patients were still on pembrolizumab while four patients discontinued therapy due to progressive disease and one due to COVID-19 infection. Half of the patients with PSA50 had both MSI-H and pathogenic alterations in BRCA1 and BRCA2 in their G360 assays. The use of liquid biopsy to identify metastatic prostate cancer patients with MSI-H is feasible in clinical practice and may overcome some of the obstacles associated with prostate cancer tumor tissue testing. The robust activity of pembrolizumab in selected patients supports the generalized testing for MSI-H.
Collapse
Affiliation(s)
- Pedro Barata
- Deming Department of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Neeraj Agarwal
- Department of Medical Oncology, University of Utah, Salt Lake City, Utah, USA
| | - Roberto Nussenzveig
- Department of Medical Oncology, University of Utah, Salt Lake City, Utah, USA
| | - Benjamin Gerendash
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Ellen Jaeger
- Deming Department of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Whitley Hatton
- Deming Department of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Elisa Ledet
- Deming Department of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Brian Lewis
- Deming Department of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Jodi Layton
- Deming Department of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Hani Babiker
- Department of Medicine, University of Arizona Arizona Cancer Center, Tucson, Arizona, USA
| | - Alan Bryce
- Department of Oncology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Rohan Garje
- Division of Hematology, Oncology, and Blood and Marrow Transplant, University of Iowa, Iowa City, Iowa, USA.,Genitourinary Oncology Program, Division of Hematology, Oncology and Blood and Marrow Transplantation, The University of Iowa, Iowa City, Iowa, USA
| | - Cy Stein
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | | | - Philip Saylor
- Department of Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Oliver Sartor
- Deming Department of Medicine, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
381
|
Sokolova AO, Shirts BH, Konnick EQ, Tsai GJ, Goulart BHL, Montgomery B, Pritchard CC, Yu EY, Cheng HH. Complexities of Next-Generation Sequencing in Solid Tumors: Case Studies. J Natl Compr Canc Netw 2020; 18:1150-1155. [PMID: 32886903 DOI: 10.6004/jnccn.2020.7569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 03/31/2020] [Indexed: 11/17/2022]
Abstract
With the promise and potential of clinical next-generation sequencing for tumor and germline testing to impact treatment and outcomes of patients with cancer, there are also risks of oversimplification, misinterpretation, and missed opportunities. These issues risk limiting clinical benefit and, at worst, perpetuating false conclusions that could lead to inappropriate treatment selection, avoidable toxicity, and harm to patients. This report presents 5 case studies illustrating challenges and opportunities in clinical next-generation sequencing interpretation and clinical application in solid tumor oncologic care. First is a case that dissects the origin of an ATM mutation as originating from a hematopoietic clone rather than the tumor. Second is a case illustrating the potential for tumor sequencing to suggest germline variants associated with a hereditary cancer syndrome. Third are 2 cases showing the potential for variant reclassification of a germline variant of uncertain significance when considered alongside family history and tumor sequencing results. Finally, we describe a case illustrating challenges with using microsatellite instability for predicting tumor response to immune checkpoint inhibitors. The common theme of the case studies is the importance of examining clinical context alongside expert review and interpretation, which together highlight an expanding role for contextual examination and multidisciplinary expert review through molecular tumor boards.
Collapse
Affiliation(s)
- Alexandra O Sokolova
- 1Department of Medicine, University of Washington.,2Clinical Research Division, Fred Hutchinson Cancer Research Center.,4Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Brian H Shirts
- 4Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Eric Q Konnick
- 4Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Ginger J Tsai
- 4Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Bernardo H L Goulart
- 1Department of Medicine, University of Washington.,2Clinical Research Division, Fred Hutchinson Cancer Research Center
| | - Bruce Montgomery
- 1Department of Medicine, University of Washington.,2Clinical Research Division, Fred Hutchinson Cancer Research Center.,4Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Colin C Pritchard
- 4Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Evan Y Yu
- 1Department of Medicine, University of Washington.,2Clinical Research Division, Fred Hutchinson Cancer Research Center
| | - Heather H Cheng
- 1Department of Medicine, University of Washington.,2Clinical Research Division, Fred Hutchinson Cancer Research Center
| |
Collapse
|
382
|
Integrated immunohistochemical and molecular analysis improves diagnosis of high-grade carcinoma in the urinary bladder of patients with prior radiation therapy for prostate cancer. Mod Pathol 2020; 33:1802-1810. [PMID: 32313185 DOI: 10.1038/s41379-020-0543-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Abstract
Prostatic adenocarcinoma and urothelial carcinoma typically demonstrate distinct morphologic and immunohistochemical features. However, high-grade prostate and urothelial carcinomas sometimes show significant morphologic and immunohistochemical overlap, which can result in misdiagnosis and mistreatment. This diagnostic dilemma is particularly acute in patients previously treated with radiation and/or hormone therapy for prostate cancer, who later present with high-grade carcinoma in the urinary bladder. To address the diagnostic utility of integrated immunohistochemical and molecular analysis in this setting, we evaluated 25 high-grade carcinomas of the bladder for which morphologic features were deemed indeterminate. Our analysis included immunohistochemistry for urothelial markers (GATA3, p63, uroplakin II), prostate markers (NKX3.1, prostate specific antigen, P501S), androgen receptor (AR) and ERG, along with molecular characterization using capture-based next generation DNA sequencing. Immunohistochemical findings were concordant with the final integrated diagnosis in 21 (84%) cases. However, in three (12%) cases, immunohistochemistry supported a diagnosis of urothelial carcinoma, but molecular analysis identified the correct diagnosis of prostate cancer based on the presence of a TMPRSS2-ERG fusion. One case remained unclassifiable even after this integrated analysis. Notably, in 1 of 21 cases, the presence of a TERT promoter mutation and the absence of a TMPRSS2-ERG fusion would typically favor a diagnosis of urothelial carcinoma, but the aggregate immunohistochemical and molecular findings instead supported a diagnosis of microsatellite unstable prostatic adenocarcinoma with deep deletion of MSH2 and MSH6. Our findings highlight the importance of considering prostatic origin in high-grade carcinoma of the urinary bladder of patients with a history of treated prostate cancer, even when the immunohistochemical findings favor urothelial carcinoma. In a subset of cases, an approach that integrates immunophenotypic and molecular data may help correctly assign site of origin and prevent misdiagnosis that can result from overreliance on any individual immunohistochemical or molecular result.
Collapse
|
383
|
Parker C, Castro E, Fizazi K, Heidenreich A, Ost P, Procopio G, Tombal B, Gillessen S. Prostate cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020; 31:1119-1134. [PMID: 32593798 DOI: 10.1016/j.annonc.2020.06.011] [Citation(s) in RCA: 586] [Impact Index Per Article: 117.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- C Parker
- Royal Marsden Hospital, Sutton, UK
| | - E Castro
- Department of Medical Oncology, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Málaga, Malaga, Spain
| | - K Fizazi
- Department of Cancer Medicine, Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - A Heidenreich
- Department of Urology, Uro-Oncology, Robot-Assisted and Specialized Urologic Surgery, University Hospital of Cologne, Cologne, Germany
| | - P Ost
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - G Procopio
- Department of Medical Oncology 1, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - B Tombal
- Institut de Recherche Clinique, Université catholique de Louvain, Brussels, Belgium
| | - S Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Faculty of Biomedical Sciences, USI, Lugano, Switzerland; Division of Cancer Medicine, University of Manchester, Manchester, UK
| |
Collapse
|
384
|
Han HJ, Li YR, Roach M, Aggarwal R. Dramatic response to combination pembrolizumab and radiation in metastatic castration resistant prostate cancer. Ther Adv Med Oncol 2020; 12:1758835920936084. [PMID: 32922519 PMCID: PMC7450451 DOI: 10.1177/1758835920936084] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/22/2020] [Indexed: 01/12/2023] Open
Abstract
Immune checkpoint inhibitors targeting PD-1 and PD-L1 have demonstrated
anti-tumor activity in several advanced solid malignancies. In
previously treated metastatic castration resistant prostate cancer
(mCRPC), a small subset of patients have a therapeutic response to
checkpoint inhibition. Those who do respond to anti-PD-1/PD-L1 therapy
have a marked, durable response to treatment, suggesting some derive
long-term benefit from immune checkpoint blockade. In other cancers,
one strategy to increase the efficacy of immune checkpoint blockade is
to combine it with a pro-immune stimulatory agent, such as radiation.
Here we present a case of a patient with heavily treated mCRPC who had
a significant tumor response to concurrent pembrolizumab and radiation
therapy to the primary prostatic mass. We review the growing evidence
supporting the use of this combination therapy in other cancers and
its potential benefit and safety in mCRPC. Our report highlights a
potential therapeutic approach that should be further investigated in
previously treated mCRPC.
Collapse
Affiliation(s)
- Harry J Han
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Yun Rose Li
- Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Mack Roach
- Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Rahul Aggarwal
- Department of Medicine, Division of Hematology/Oncology, UCSF Helen Diller Family Comprehensive Cancer Center, 550 16th Street, Box 3211, San Francisco, CA, 94158, USA
| |
Collapse
|
385
|
Moss EL, Gorsia DN, Collins A, Sandhu P, Foreman N, Gore A, Wood J, Kent C, Silcock L, Guttery DS. Utility of Circulating Tumor DNA for Detection and Monitoring of Endometrial Cancer Recurrence and Progression. Cancers (Basel) 2020; 12:E2231. [PMID: 32785174 PMCID: PMC7463944 DOI: 10.3390/cancers12082231] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
Despite the increasing incidence of endometrial cancer (EC) worldwide and the poor overall survival of patients who recur, no reliable biomarker exists for detecting and monitoring EC recurrence and progression during routine follow-up. Circulating tumor DNA (ctDNA) is a sensitive method for monitoring cancer activity and stratifying patients that are likely to respond to therapy. As a pilot study, we investigated the utility of ctDNA for detecting and monitoring EC recurrence and progression in 13 patients, using targeted next-generation sequencing (tNGS) and personalized ctDNA assays. Using tNGS, at least one somatic mutation at a variant allele frequency (VAF) > 20% was detected in 69% (9/13) of patient tumors. The four patients with no detectable tumor mutations at >20% VAF were whole exome sequenced, with all four harboring mutations in genes not analyzed by tNGS. Analysis of matched and longitudinal plasma DNA revealed earlier detection of EC recurrence and progression and dynamic kinetics of ctDNA levels reflecting treatment response. We also detected acquired high microsatellite instability (MSI-H) in ctDNA from one patient whose primary tumor was MSI stable. Our study suggests that ctDNA analysis could become a useful biomarker for early detection and monitoring of EC recurrence. However, further research is needed to confirm these findings and to explore their potential implications for patient management.
Collapse
Affiliation(s)
- Esther L. Moss
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE2 7LX, UK; (D.N.G.); (A.C.); (P.S.); (N.F.)
- Department of Gynaecological Oncology, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester LE5 4PW, UK; (A.G.); (J.W.); (C.K.)
| | - Diviya N. Gorsia
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE2 7LX, UK; (D.N.G.); (A.C.); (P.S.); (N.F.)
| | - Anna Collins
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE2 7LX, UK; (D.N.G.); (A.C.); (P.S.); (N.F.)
| | - Pavandeep Sandhu
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE2 7LX, UK; (D.N.G.); (A.C.); (P.S.); (N.F.)
| | - Nalini Foreman
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE2 7LX, UK; (D.N.G.); (A.C.); (P.S.); (N.F.)
| | - Anupama Gore
- Department of Gynaecological Oncology, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester LE5 4PW, UK; (A.G.); (J.W.); (C.K.)
| | - Joey Wood
- Department of Gynaecological Oncology, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester LE5 4PW, UK; (A.G.); (J.W.); (C.K.)
| | - Christopher Kent
- Department of Gynaecological Oncology, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester LE5 4PW, UK; (A.G.); (J.W.); (C.K.)
| | - Lee Silcock
- Nonacus Limited, Birmingham Research Park, Birmingham B15 2SQ, UK;
| | - David S. Guttery
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE2 7LX, UK; (D.N.G.); (A.C.); (P.S.); (N.F.)
| |
Collapse
|
386
|
Montgomery B, Rettig M, Kasten J, Muralidhar S, Myrie K, Ramoni R. The Precision Oncology Program for Cancer of the Prostate (POPCaP) Network: A Veterans Affairs/Prostate Cancer Foundation Collaboration. Fed Pract 2020; 37:S48-S53. [PMID: 32908353 PMCID: PMC7473728 DOI: 10.12788/fp.0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The promise of precision oncology only can be realized when genetic alterations are identified that can be lever-aged to improve response and minimize toxicity. Identifying those alterations as efficiently as possible and then giving patients access to targeted therapy and clinical study requires a comprehensive strategy across health care systems. OBSERVATIONS The US Department of Veterans Affairs (VA) and Prostate Cancer Foundation have established a network of VA centers to help develop best practices for precision oncology for the treatment of veterans with advanced prostate cancer. This article describes the genesis and structure of this network and its potential for contributing to care and research in the VA and the health care system as a whole. CONCLUSIONS The Precision Oncology Program for Cancer of the Prostate network and its partnership with VA clinical and research efforts is anticipated to provide important insights into barriers and solutions to the implementation of precision oncology for prostate cancer across the VA.
Collapse
Affiliation(s)
- Bruce Montgomery
- is an Oncologist and is Network Director at VA Puget Sound Health Care System in Seattle, Washington. is an Oncologist at the Greater Los Angeles Health Care System and a Professor of Medicine at the University of California, Los Angeles. is Program Director, Million Veteran Program. is Scientific Program Manager of Oncology and is Chief Research and Development Officer, all in the Office of Research and Development, Veterans Health Administration. Bruce Montgomery is a Professor of Medicine at the University of Washington in Seattle
| | - Matthew Rettig
- is an Oncologist and is Network Director at VA Puget Sound Health Care System in Seattle, Washington. is an Oncologist at the Greater Los Angeles Health Care System and a Professor of Medicine at the University of California, Los Angeles. is Program Director, Million Veteran Program. is Scientific Program Manager of Oncology and is Chief Research and Development Officer, all in the Office of Research and Development, Veterans Health Administration. Bruce Montgomery is a Professor of Medicine at the University of Washington in Seattle
| | - Jesse Kasten
- is an Oncologist and is Network Director at VA Puget Sound Health Care System in Seattle, Washington. is an Oncologist at the Greater Los Angeles Health Care System and a Professor of Medicine at the University of California, Los Angeles. is Program Director, Million Veteran Program. is Scientific Program Manager of Oncology and is Chief Research and Development Officer, all in the Office of Research and Development, Veterans Health Administration. Bruce Montgomery is a Professor of Medicine at the University of Washington in Seattle
| | - Sumitra Muralidhar
- is an Oncologist and is Network Director at VA Puget Sound Health Care System in Seattle, Washington. is an Oncologist at the Greater Los Angeles Health Care System and a Professor of Medicine at the University of California, Los Angeles. is Program Director, Million Veteran Program. is Scientific Program Manager of Oncology and is Chief Research and Development Officer, all in the Office of Research and Development, Veterans Health Administration. Bruce Montgomery is a Professor of Medicine at the University of Washington in Seattle
| | - Kenute Myrie
- is an Oncologist and is Network Director at VA Puget Sound Health Care System in Seattle, Washington. is an Oncologist at the Greater Los Angeles Health Care System and a Professor of Medicine at the University of California, Los Angeles. is Program Director, Million Veteran Program. is Scientific Program Manager of Oncology and is Chief Research and Development Officer, all in the Office of Research and Development, Veterans Health Administration. Bruce Montgomery is a Professor of Medicine at the University of Washington in Seattle
| | - Rachel Ramoni
- is an Oncologist and is Network Director at VA Puget Sound Health Care System in Seattle, Washington. is an Oncologist at the Greater Los Angeles Health Care System and a Professor of Medicine at the University of California, Los Angeles. is Program Director, Million Veteran Program. is Scientific Program Manager of Oncology and is Chief Research and Development Officer, all in the Office of Research and Development, Veterans Health Administration. Bruce Montgomery is a Professor of Medicine at the University of Washington in Seattle
| |
Collapse
|
387
|
Advanced Prostate Cancer: Treatment Advances and Future Directions. Trends Cancer 2020; 6:702-715. [DOI: 10.1016/j.trecan.2020.04.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 11/18/2022]
|
388
|
Kim TJ, Koo KC. Current Status and Future Perspectives of Checkpoint Inhibitor Immunotherapy for Prostate Cancer: A Comprehensive Review. Int J Mol Sci 2020; 21:E5484. [PMID: 32751945 PMCID: PMC7432105 DOI: 10.3390/ijms21155484] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
The clinical spectrum of prostate cancer (PCa) varies from castration-naive to metastatic castration-resistant disease. Despite the administration of androgen synthesis inhibitors and chemotherapy regimens for castration-resistant prostate cancer, the treatment options for this entity are limited. The utilization of the immune system against cancer cells shows potential as a therapeutic modality for various solid tumors and hematologic malignancies. With technological advances over the last decade, immunotherapy has become an integral treatment modality for advanced solid tumors. The feasibility of immunotherapy has shown promise for patients with PCa, and with advances in molecular diagnostic platforms and our understanding of immune mechanisms, immunotherapy is reemerging as a potential treatment modality for PCa. Various combinations of individualized immunotherapy and immune checkpoint blockers with androgen receptor-targeted therapies and conventional cytotoxic agents show promise. This article will review the current status of immunotherapy, including new discoveries and precision approaches to PCa, and discuss future directions in the continuously evolving landscape of immunotherapy.
Collapse
Affiliation(s)
- Tae Jin Kim
- Department of Urology, C.H.A. Bundang Medical Center, University College of Medicine, Seongnam 13496, Korea;
| | - Kyo Chul Koo
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Korea
| |
Collapse
|
389
|
Nickols NG, Ganapathy E, Nguyen C, Kane N, Lin L, Diaz-Perez S, Nazarian R, Mathis C, Felix C, Basehart V, Zomorodian N, Kwak J, Kishan AU, King CR, Kupelian PA, Rettig MB, Steinberg ML, Cao M, Knudsen BS, Chu FI, Romero T, Elashoff D, Reiter RE, Schaue D. The intraprostatic immune environment after stereotactic body radiotherapy is dominated by myeloid cells. Prostate Cancer Prostatic Dis 2020; 24:135-139. [PMID: 32647353 PMCID: PMC7794088 DOI: 10.1038/s41391-020-0249-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/08/2020] [Accepted: 06/30/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND: Hundreds of ongoing clinical trials combine radiation therapy, mostly delivered as stereotactic body radiotherapy (SBRT), with immune checkpoint blockade. However, our understanding of the effect of radiotherapy on the intratumoral immune balance is inadequate, hindering the optimal design of trials that combine radiation therapy with immunotherapy. Our objective was to characterize the intratumoral immune balance of the malignant prostate after SBRT in patients. METHODS: 16 patients with high-risk, non-metastatic prostate cancer at comparable Gleason Grade disease underwent radical prostatectomy with (n=9) or without (n=7) neoadjuvant SBRT delivered in 3 fractions of 8 Gy over 5 days completed 2 weeks before surgery. Freshly resected prostate specimens were processed to obtain single-cell suspensions, and immune-phenotyped for major lymphoid and myeloid cell subsets by staining with 2 separate 14-antibody panels and multicolor flow cytometry analysis. RESULTS: Malignant prostates two weeks after SBRT had an immune infiltrate dominated by myeloid cells, whereas malignant prostates without preoperative treatment were more lymphoid-biased (myeloid CD45+ cells 48.4 ± 19.7% vs 25.4 ± 7.0%; adjusted p value=0.11; and CD45+ lymphocytes 51.6 ± 19.7% vs 74.5 ± 7.0%; p=0.11; CD3+ T cells 35.2 ± 23.8% vs 60.9 ± 9.7%; p=0.12; mean±SD). CONCLUSION: SBRT drives a significant lymphoid to myeloid shift in the prostate tumor immune infiltrate. This may be of interest when combining SBRT with immunotherapies, particularly in prostate cancer.
Collapse
Affiliation(s)
- Nicholas G Nickols
- Radiation Oncology at UCLA, Los Angeles, CA, USA.,Urology at UCLA, Los Angeles, CA, USA.,VA Greater Los Angeles Healthcare System, Radiation Therapy Service, Los Angeles, CA, USA.,UCLA Jonsson Compressive Cancer Center, Los Angeles, CA, USA
| | | | | | | | - Lin Lin
- Urology at UCLA, Los Angeles, CA, USA
| | | | | | | | - Care Felix
- Radiation Oncology at UCLA, Los Angeles, CA, USA
| | | | | | - Jae Kwak
- Urology at UCLA, Los Angeles, CA, USA
| | - Amar U Kishan
- Radiation Oncology at UCLA, Los Angeles, CA, USA.,Urology at UCLA, Los Angeles, CA, USA.,UCLA Jonsson Compressive Cancer Center, Los Angeles, CA, USA
| | | | | | - Matthew B Rettig
- Urology at UCLA, Los Angeles, CA, USA.,UCLA Jonsson Compressive Cancer Center, Los Angeles, CA, USA
| | - Michael L Steinberg
- Radiation Oncology at UCLA, Los Angeles, CA, USA.,UCLA Jonsson Compressive Cancer Center, Los Angeles, CA, USA
| | - Minsong Cao
- Radiation Oncology at UCLA, Los Angeles, CA, USA
| | - Beatrice S Knudsen
- Pathology and Laboratory Medicine and Biomedical Sciences at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Fang-I Chu
- Radiation Oncology at UCLA, Los Angeles, CA, USA
| | - Tahmineh Romero
- Division of General Internal Medicine and Health Services Research at UCLA, Los Angeles, CA, USA
| | - David Elashoff
- UCLA Jonsson Compressive Cancer Center, Los Angeles, CA, USA.,Division of General Internal Medicine and Health Services Research at UCLA, Los Angeles, CA, USA
| | - Robert E Reiter
- Urology at UCLA, Los Angeles, CA, USA.,UCLA Jonsson Compressive Cancer Center, Los Angeles, CA, USA
| | - Dörthe Schaue
- Radiation Oncology at UCLA, Los Angeles, CA, USA. .,UCLA Jonsson Compressive Cancer Center, Los Angeles, CA, USA.
| |
Collapse
|
390
|
Camrelizumab in advanced or metastatic solid tumour patients with DNA mismatch repair deficient or microsatellite instability high: an open-label prospective pivotal trial. J Cancer Res Clin Oncol 2020; 146:2651-2657. [PMID: 32623573 DOI: 10.1007/s00432-020-03251-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/07/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Patients with DNA mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) cancers are prone to response to programmed cell death-1 (PD-1) checkpoint inhibitors. Therefore, we explored the efficacy and safety of a PD-1 checkpoint inhibitor camrelizumab in advanced or metastatic solid tumour with dMMR/MSI-H. METHODS Patients with dMMR/MSI-H advanced or metastatic solid tumours who had received at least one line of prior systemic chemotherapy were recruited. Camrelizumab was given intravenously 200 mg every 2-week treatment cycle. The primary endpoint was objective response rate according to Response Evaluation Criteria in Solid Tumours v1.1. RESULTS Twelve patients were enrolled. As data cutoff, eight patients (66.7%, 95% CI 34.9-90.1) achieved objective response. Disease control rate reached 100% (95% CI 73.5-100). Progression-free survival rate at 12 months was 83.3% (95% CI 48.2-95.6), and overall survival rate at 12 months was 90% (95% CI 47.3-98.5). The most common treatment-related adverse events were reactive cutaneous capillary endothelial proliferation (100%), increased alanine aminotransferase (41.7%), and increased aspartate aminotransferase (41.7%). CONCLUSIONS Camrelizumab provided durable objective response and disease control in pre-treated patients with dMMR/MSI-H advanced or metastatic solid tumour, being a promising treatment option for these patients.
Collapse
|
391
|
Jiao XD, Zhang XC, Qin BD, Liu D, Liu L, Ni JJ, Ning ZY, Chen LX, Zhu LJ, Qin SB, Ying SP, Chen XQ, Li AJ, Hou T, Han-Zhang H, Ye J, Zheng J, Chuai S, Zang YS. Tumor mutation burden in Chinese cancer patients and the underlying driving pathways of high tumor mutation burden across different cancer types. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:860. [PMID: 32793704 DOI: 10.21037/atm-20-3807] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Tumor mutation burden (TMB) has an important association with immunotherapy responses. TMB in the Chinese population has not been well established. Finding differences between the Chinese and Caucasian populations and elucidating the underlying biological mechanisms of high TMB might help develop more precise and effective means for TMB and immunotherapy response prediction. Methods Chinese cancer patients fresh tissue (n=2,177), formalin-fixed, paraffin-embed (FFPE) specimens (n=3,294), and pleural fluid (n=189) were profiled using a 295- or 520-gene next-generation sequencing (NGS) panel. The association of the TMB status with a series of molecular features and biological pathways was determined using bootstrapping. Results TMB, measured by 295- or 520-cancer-related gene panels, was correlated with whole-exome sequencing (WES) TMB based on the in silico simulation in The Cancer Genome Atlas cohort. The median TMB of our data was slightly higher than that from the Foundation Medicine Inc. (FMI) dataset. TMB was also slightly different within the same cancer type between the Chinese and Caucasian population. We discovered that the underlying pathways of TMB status varied greatly and sometimes had an opposite association with TMB across different cancer types. Moreover, we developed a 23-gene and a 16-gene signature to predict TMB prediction for lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), respectively, indicating a histology-specific mechanism for driving high-TMB in lung cancer. Conclusions TMB varies among different ethnic populations. Our findings extend the knowledge of the underlying biological mechanisms for high TMB and might be helpful for developing more precise and accessible TMB assessment panels and algorithms in more cancer types.
Collapse
Affiliation(s)
- Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiao-Chun Zhang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Dong Liu
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Liang Liu
- Departments of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jian-Jiao Ni
- Departments of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhou-Yu Ning
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ling-Xiang Chen
- Department of Internal Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Liang-Jun Zhu
- Department of Internal Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Song-Bing Qin
- Department of Tumor Radiotherapy, The First Affiliated Hospital of Suzhou University, Suzhou, China
| | - Shen-Peng Ying
- Department of Radiotherapy, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, China
| | - Xue-Qin Chen
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ai-Jun Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Ting Hou
- Burning Rock Biotech, Guangzhou, China
| | | | - Junyi Ye
- Burning Rock Biotech, Guangzhou, China
| | | | | | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
392
|
Alqaisi HA, Al-Ezzi E, Hansen AR. Biomarkers of response to immune checkpoint inhibitors for metastatic castration resistant prostate cancer: looking for the needle in the haystack. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:894. [PMID: 32793738 PMCID: PMC7396782 DOI: 10.21037/atm.2020.03.78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 03/28/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Husam A Alqaisi
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Esmail Al-Ezzi
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Aaron R Hansen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
393
|
Markowski MC, Antonarakis ES. PARP inhibitors in prostate cancer: time to narrow patient selection? Expert Rev Anticancer Ther 2020; 20:523-526. [PMID: 32521178 PMCID: PMC9774050 DOI: 10.1080/14737140.2020.1781622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Mark C. Markowski
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Emmanuel S. Antonarakis
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA,Urology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|
394
|
U Gandhy S, Madan RA, Aragon-Ching JB. The immunotherapy revolution in genitourinary malignancies. Immunotherapy 2020; 12:819-831. [PMID: 32594815 DOI: 10.2217/imt-2020-0054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy and therapeutic cancer vaccines have continued to demonstrate survival benefit and durable clinical response in patients with renal cell cancer, prostate cancer and bladder cancer, with limited responses in testicular cancer. The role of immunotherapy in combination with chemotherapy or other targeted therapies in the neo-adjuvant, adjuvant and metastatic setting is actively being explored. We describe the current immunotherapy-related treatment modalities approved for genitourinary cancers, focusing on immune checkpoint inhibitors, vaccines and other modalities, and highlight ongoing studies involving immunotherapy in these cancer types.
Collapse
Affiliation(s)
- Shruti U Gandhy
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
395
|
Abstract
For decades, the treatment of advanced prostate cancer was mainly based on the manipulation of the androgen receptor-controlled proliferation pathway. Chemotherapy only played an additional important role with the advent of taxanes. The progress in translational research in recent years has led to innovations in the therapeutic environment. With the decoding of the homologous repair deficiency (HRD) machinery and its ability to be influenced by PARP inhibitors, targeted therapies moved into the therapeutic focus for selected patients. The first positive phase III study for PARP inhibitors is already available. In addition, immunotherapy for the treatment of prostate cancer, which is now widely used in oncology, is also making progress; both checkpoint inhibitors and bispecific antibodies have shown clinically useful activities. Cellular therapies such as CAR T cells, which are directed against prostate-specific membrane antigen (PSMA), are still at an early stage of development. In this review, the authors provide a summary of the basic principles and clinical development of these new therapies.
Collapse
|
396
|
Crippa A, De Laere B, Discacciati A, Larsson B, Connor JT, Gabriel EE, Thellenberg C, Jänes E, Enblad G, Ullen A, Hjälm-Eriksson M, Oldenburg J, Ost P, Lindberg J, Eklund M, Grönberg H. The ProBio trial: molecular biomarkers for advancing personalized treatment decision in patients with metastatic castration-resistant prostate cancer. Trials 2020; 21:579. [PMID: 32586393 PMCID: PMC7318749 DOI: 10.1186/s13063-020-04515-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Multiple therapies exist for patients with metastatic castration-resistant prostate cancer (mCRPC). However, their improvement on progression-free survival (PFS) remains modest, potentially explained by tumor molecular heterogeneity. Several prognostic molecular biomarkers have been identified for mCRPC that may have predictive potential to guide treatment selection and prolong PFS. We designed a platform trial to test this hypothesis. METHODS The Prostate-Biomarker (ProBio) study is a multi-center, outcome-adaptive, multi-arm, biomarker-driven platform trial for tailoring treatment decisions for men with mCRPC. Treatment decisions in the experimental arms are based on biomarker signatures defined as mutations in certain genes/pathways suggested in the scientific literature to be important for treatment response in mCRPC. The biomarker signatures are determined by targeted sequencing of circulating tumor and germline DNA using a panel specifically designed for mCRPC. DISCUSSION Patients are stratified based on the sequencing results and randomized to either current clinical practice (control), where the treating physician decides treatment, or to molecularly driven treatment selection based on the biomarker profile. Outcome-adaptive randomization is implemented to early identify promising treatments for a biomarker signature. Biomarker signature-treatment combinations graduate from the platform when they demonstrate 85% probability of improving PFS compared to the control arm. Graduated combinations are further evaluated in a seamless confirmatory trial with fixed randomization. The platform design allows for new drugs and biomarkers to be introduced in the study. CONCLUSIONS The ProBio design allows promising treatment-biomarker combinations to quickly graduate from the platform and be confirmed for rapid implementation in clinical care. TRIAL REGISTRATION ClinicalTrials.gov Identifier NCT03903835. Date of registration: April 4, 2019. Status: Recruiting.
Collapse
Affiliation(s)
- Alessio Crippa
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Bram De Laere
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Andrea Discacciati
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Berit Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jason T Connor
- University of Central Florida College of Medicine, Orlando, FL, USA
- Confluence Stat LLC, Orlando, FL, USA
| | - Erin E Gabriel
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Camilla Thellenberg
- Department of Radiation Sciences and Oncology, Umeå University, Umeå, Sweden
| | - Elin Jänes
- Länssjukhuset Sundsvall Härnösand, Sundsvall, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology, Uppsala Universitet, Uppsala, Sweden
| | - Anders Ullen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Oldenburg
- Division of Medicine, University of Oslo, Oslo, Norway
| | - Piet Ost
- Department of Radiotherapy and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
397
|
Aurilio G, Cimadamore A, Santoni M, Nolè F, Scarpelli M, Massari F, Lopez-Beltran A, Cheng L, Montironi R. New Frontiers in Prostate Cancer Treatment: Are We Ready for Drug Combinations with Novel Agents? Cells 2020; 9:cells9061522. [PMID: 32580469 PMCID: PMC7349416 DOI: 10.3390/cells9061522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
Medical treatment for metastatic castration-resistant prostate cancer (mCRPC) patients has progressively been evolving from a nonspecific clinical approach to genomics-oriented therapies. The scientific community is in fact increasingly focusing on developing DNA damage repair (DDR) defect-driven novel molecules, both as single-agent therapy and in combined treatment strategies. Accordingly, research is under way into combined drug therapies targeting different pathways, e.g. androgen receptor signaling (ARS) and poly (adenosine diphosphate [ADP]-ribose) polymerase (PARP) enzymes, immune checkpoint (IC) and PARP, IC, and ARS, and prostate-specific membrane antigen (PSMA). In an attempt to formulate evolving treatment paradigms in mCRPC patients, here we selected clinical research into patients undergoing therapies with emerging molecules, with particular emphasis towards PARP-, IC-, and PSMA-inhibitors. In order to focus on those molecules and drug combinations most likely to be translated into routine clinical care in the near future, we selected only those clinical studies currently recruiting patients. A PubMed search focusing on the keywords “prostate cancer”, “metastatic castration-resistant prostate cancer”, “DDR pathways”, “ARS inhibitors”, “PARP inhibitors”, “IC inhibitors”, “PSMA-targeting agents”, and “drug combinations” was performed.
Collapse
Affiliation(s)
- Gaetano Aurilio
- Medical Oncology Division of Urogenital and Head and Neck Tumours, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
- Correspondence: ; Tel.: +390257489502; Fax: +390294379234
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62012 Macerata, Italy;
| | - Franco Nolè
- Medical Oncology Division of Urogenital and Head and Neck Tumours, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Francesco Massari
- Division of Oncology, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy;
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| |
Collapse
|
398
|
Cimadamore A, Aurilio G, Nolé F, Massari F, Scarpelli M, Santoni M, Lopez-Beltran A, Cheng L, Montironi R. Update on Circulating Tumor Cells in Genitourinary Tumors with Focus on Prostate Cancer. Cells 2020; 9:E1495. [PMID: 32575429 PMCID: PMC7348874 DOI: 10.3390/cells9061495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Current developments in the treatment of genitourinary tumors underline the unmet clinical need for biomarkers to improve decision-making in a challenging clinical setting. The detection of circulating tumor cells (CTCs) has become one of the most exciting and important new approaches to identifying biomarkers at different stages of disease in a non-invasive way. Potential applications of CTCs include monitoring treatment efficacy and early detection of progression, selecting tailored therapies, as well as saving treatment costs. However, despite the promising implementation of CTCs in a clinical scenario, the isolation and characterization of these cells for molecular studies remain expensive with contemporary platforms, and significant technical challenges still need to be overcome. This updated, critical review focuses on the state of CTCs in patients with genitourinary tumor with focus on prostate cancer, discussing technical issues, main clinical results and hypothesizing potential future perspectives in clinical scenarios.
Collapse
Affiliation(s)
- Alessia Cimadamore
- Section of Pathological Anatomy, Faculty of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (M.S.); (R.M.)
| | - Gaetano Aurilio
- Department of Medical Oncology, Division of Urogenital and Head and Neck Tumours, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (G.A.); (F.N.)
| | - Franco Nolé
- Department of Medical Oncology, Division of Urogenital and Head and Neck Tumours, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (G.A.); (F.N.)
| | - Francesco Massari
- Division of Oncology, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy;
| | - Marina Scarpelli
- Section of Pathological Anatomy, Faculty of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (M.S.); (R.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN 462020, USA;
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Faculty of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (M.S.); (R.M.)
| |
Collapse
|
399
|
Couñago F, López-Campos F, Díaz-Gavela AA, Almagro E, Fenández-Pascual E, Henríquez I, Lozano R, Linares Espinós E, Gómez-Iturriaga A, de Velasco G, Quintana Franco LM, Rodríguez-Melcón I, López-Torrecilla J, Spratt DE, Guerrero LL, Martínez-Salamanca JI, del Cerro E. Clinical Applications of Molecular Biomarkers in Prostate Cancer. Cancers (Basel) 2020; 12:1550. [PMID: 32545454 PMCID: PMC7352850 DOI: 10.3390/cancers12061550] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
There is clinically relevant molecular heterogeneity in prostate cancer (PCa), but this biological diversity has had only a minimal impact on clinical practice. Treatment outcomes in patients with localised PCa are often highly variable, even among patients stratified to the same risk group or disease state based on standard clinical and pathological parameters. In recent years, the development of gene panels has provided valuable data on the differential expression of genes in patients with PCa. Nevertheless, there is an urgent need to identify and validate prognostic and predictive biomarkers that can be applied across clinical scenarios, ranging from localised disease to metastatic castration-resistant PCa. The availability of such tools would allow for precision medicine to finally reach PCa patients. In this review, we evaluate current data on molecular biomarkers for PCa, with an emphasis on the biomarkers and gene panels with the most robust evidence to support their application in routine clinical practice.
Collapse
Affiliation(s)
- Felipe Couñago
- Radiation Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain; (A.A.D.-G.); (L.L.G.); (E.d.C.)
- Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Clinical Department, Faculty of Biomedicine. Universidad Europea de Madrid, 28670 Madrid, Spain
| | | | - Ana Aurora Díaz-Gavela
- Radiation Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain; (A.A.D.-G.); (L.L.G.); (E.d.C.)
- Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Clinical Department, Faculty of Biomedicine. Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Elena Almagro
- Medical Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain;
| | - Esaú Fenández-Pascual
- Lyx Institute of Urology, Universidad Francisco de Vitoria, 28006 Madrid, Spain; (E.F.-P.); (E.L.E.)
- Department of Urology, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | - Iván Henríquez
- Radiation Oncology, Hospital Universitario Sant Joan, 43204 Reus, Spain;
| | - Rebeca Lozano
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, 28029 Madrid, Spain;
- Genitourinary Cancer Traslational Research Group, Institute of Biomedical Research, 29010 Málaga, Spain
| | - Estefanía Linares Espinós
- Lyx Institute of Urology, Universidad Francisco de Vitoria, 28006 Madrid, Spain; (E.F.-P.); (E.L.E.)
- Department of Urology, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | | | | | | | - Ignacio Rodríguez-Melcón
- Radiation Oncology, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Spain;
| | - José López-Torrecilla
- Radiation Oncology-ERESA, Hospital General Universitario de Valencia, 46014 Valencia, Spain;
| | - Daniel E. Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Luis Leonardo Guerrero
- Radiation Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain; (A.A.D.-G.); (L.L.G.); (E.d.C.)
- Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Clinical Department, Faculty of Biomedicine. Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Juan Ignacio Martínez-Salamanca
- Lyx Institute of Urology, Universidad Francisco de Vitoria, 28006 Madrid, Spain; (E.F.-P.); (E.L.E.)
- Department of Urology, Hospital Universitario Puerta de Hierro, 28222 Madrid, Spain
| | - Elia del Cerro
- Radiation Oncology, Hospital Universitario Quirónsalud Madrid, 28223 Madrid, Spain; (A.A.D.-G.); (L.L.G.); (E.d.C.)
- Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Clinical Department, Faculty of Biomedicine. Universidad Europea de Madrid, 28670 Madrid, Spain
| |
Collapse
|
400
|
Poor Clinical Outcomes and Immunoevasive Contexture in Intratumoral IL-10-Producing Macrophages Enriched Gastric Cancer Patients. Ann Surg 2020; 275:e626-e635. [PMID: 32541216 DOI: 10.1097/sla.0000000000004037] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the clinical significance of IL-10 tumor-associated macrophages (TAMs) in gastric cancer. BACKGROUND Due to the plasticity and diversity of TAMs, it is necessary to phenotypically and functionally classify subsets of TAMs to better understand the critical role of TAMs in cancer progression. TAMs expressing interleukin-10 (IL-10) have been found to facilitate immune evasion in many malignancies, but the role of IL-10 TAMs in gastric cancer remains obscure. METHODS Four hundred and sixty-eight tumor tissue microarray specimens, 52 fresh tumor tissue samples of gastric cancer patients from Zhongshan Hospital, and data of 298 gastric cancer patients from the Cancer Genome Atlas (TCGA) were analyzed. IL-10 TAM level and immune contexture were examined by CIBERSORT, immunohistochemistry, and flow cytometry. Clinical outcomes were analyzed by Kaplan-Meier curves and Cox model. RESULTS Gastric cancer patients with high IL-10 TAM infiltration exhibited poor prognosis and inferior therapeutic responsiveness to fluorouracil-based adjuvant chemotherapy. IL-10 TAM infiltration yielded an immunoevasive tumor microenvironment featured by regulatory T cell infiltration and CD8 T cell dysfunction. The combinational analysis of IL-10 TAM and CD8 T cell infiltration stratified patients into distinct risk groups with different clinical outcomes. Moreover, IL-10 TAM infiltration was correlated with tumor-intrinsic characteristics including EBV status, PD-L1 expression, and genome stability in gastric cancer. CONCLUSIONS This study revealed that IL-10 TAMs might drive an immunoevasive microenvironment and determine poor prognosis and inferior therapeutic responsiveness to fluorouracil-based adjuvant chemotherapy, indicating IL-10 TAMs could be applied as a potential target for immunotherapeutic approach in gastric cancer.
Collapse
|