1
|
Schoeps B, Lauer UM, Elbers K. Deciphering permissivity of human tumor ecosystems to oncolytic viruses. Oncogene 2025; 44:1069-1077. [PMID: 40148688 PMCID: PMC11996678 DOI: 10.1038/s41388-025-03357-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/10/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Effective cancer therapy involves initiation of a tumor-specific immune response. Consequently, the interest in oncolytic viruses (OV) capable of triggering immunogenic cell death has sparked in recent years. However, the common use of pre-clinical models that fail to mirror patient tumor ecosystems (TES) hinders clinical translation. Here, we provide a condensed view on the intricate interplay between several aspects of TES and OV action and discuss these considerations in the view of recently developed pre-clinical human model systems. Given the urgent demand for innovative cancer treatments, the purpose of this review is to highlight the so-far overlooked complex impact of the tumor microenvironment (TME) on OV permissivity, with the intent to provide a foundation for future, more effective pre-clinical studies.
Collapse
Affiliation(s)
| | - Ulrich M Lauer
- Department of Medical Oncology and Pneumology, Virotherapy Center Tübingen (VCT), Medical University Hospital, Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Tübingen, Germany
| | | |
Collapse
|
2
|
Sederman C, Yang CH, Cortes-Sanchez E, Di Sera T, Huang X, Scherer SD, Zhao L, Chu Z, White ER, Atkinson A, Wagstaff J, Varley KE, Lewis MT, Qiao Y, Welm BE, Welm AL, Marth GT. A precision oncology-focused deep learning framework for personalized selection of cancer therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628190. [PMID: 39763776 PMCID: PMC11702554 DOI: 10.1101/2024.12.12.628190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Precision oncology matches tumors to targeted therapies based on the presence of actionable molecular alterations. However, most tumors lack actionable alterations, restricting treatment options to cytotoxic chemotherapies for which few data-driven prioritization strategies currently exist. Here, we report an integrated computational/experimental treatment selection approach applicable for both chemotherapies and targeted agents irrespective of actionable alterations. We generated functional drug response data on a large collection of patient-derived tumor models and used it to train ScreenDL, a novel deep learning-based cancer drug response prediction model. ScreenDL leverages the combination of tumor omic and functional drug screening data to predict the most efficacious treatments. We show that ScreenDL accurately predicts response to drugs with diverse mechanisms, outperforming existing methods and approved biomarkers. In our preclinical study, this approach achieved superior clinical benefit and objective response rates in breast cancer patient-derived xenografts, suggesting that testing ScreenDL in clinical trials may be warranted.
Collapse
Affiliation(s)
- Casey Sederman
- Eccles Institute of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Chieh-Hsiang Yang
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Emilio Cortes-Sanchez
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Tony Di Sera
- Eccles Institute of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Xiaomeng Huang
- Eccles Institute of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Sandra D Scherer
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Ling Zhao
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Zhengtao Chu
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Eliza R White
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Aaron Atkinson
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Jadon Wagstaff
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Katherine E Varley
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Michael T Lewis
- Departments of Molecular and Cellular Biology and Radiology. Lester and Sue Smith Breast Center. Dan L Duncan Comprehensive Cancer Center. Baylor College of Medicine, Houston, Texas, USA
| | - Yi Qiao
- Department of Biomedical Informatics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Bryan E Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Surgery, University of Utah, Salt Lake City, UT, USA
| | - Alana L Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Gabor T Marth
- Eccles Institute of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
3
|
Montagner A, Arleo A, Suzzi F, D’Assoro AB, Piscaglia F, Gramantieri L, Giovannini C. Notch Signaling and PD-1/PD-L1 Interaction in Hepatocellular Carcinoma: Potentialities of Combined Therapies. Biomolecules 2024; 14:1581. [PMID: 39766289 PMCID: PMC11674819 DOI: 10.3390/biom14121581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy has shown significant improvement in the survival of patients with hepatocellular carcinoma (HCC) compared to TKIs as first-line treatment. Unfortunately, approximately 30% of HCC exhibits intrinsic resistance to ICIs, making new therapeutic combinations urgently needed. The dysregulation of the Notch signaling pathway observed in HCC can affect immune cell response, reducing the efficacy of cancer immunotherapy. Here, we provide an overview of how Notch signaling regulates immune responses and present the therapeutic rationale for combining Notch signaling inhibition with ICIs to improve HCC treatment. Moreover, we propose using exosomes as non-invasive tools to assess Notch signaling activation in hepatic cancer cells, enabling accurate stratification of patients who can benefit from combined strategies.
Collapse
Affiliation(s)
- Annapaola Montagner
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55902, USA;
| | - Andrea Arleo
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
| | - Fabrizia Suzzi
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
| | - Antonino B. D’Assoro
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55902, USA;
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Catia Giovannini
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (A.A.); (F.S.); (F.P.); (C.G.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| |
Collapse
|
4
|
Zhou K, Du L, Ding R, Xu L, Shi S, Wang S, Wang Z, Zhang G, He G, Zhao Z, Tang BZ. Photocatalytic therapy via photoinduced redox imbalance in biological system. Nat Commun 2024; 15:10551. [PMID: 39632877 PMCID: PMC11618361 DOI: 10.1038/s41467-024-55060-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024] Open
Abstract
Redox balance is essential for sustaining normal physiological metabolic activities of life. In this study, we present a photocatalytic system to perturb the balance of NADH/NAD+ in oxygen-free conditions, achieving photocatalytic therapy to cure anaerobic bacterial infected periodontitis. Under light irradiation, the catalyst TBSMSPy+ can bind bacterial DNA and initiate the generation of radical species through a multi-step electron transfer process. It catalyzes the conversion from NADH to NAD+ (the turnover frequency up to 60.7 min-1), inhibits ATP synthesis, disrupts the energy supply required for DNA replication, and successfully accomplishes photocatalytic sterilization in an oxygen-free environment. The catalyst participates in the redox reaction, interfering with the balance of NADH/NAD+ contents under irradiation, so we termed this action as photoinduced redox imbalance. Additionally, animal experiments in male rats also validate that the TBSMSPy+ could effectively catalyze the NADH oxidation, suppress metabolism and stimulate osteogenesis. Our research substantiates the concept of photoinduced redox imbalance and the application of photocatalytic therapy, further advocating the development of such catalyst based on photoinduced redox imbalance strategy for oxygen-free phototherapy.
Collapse
Grants
- 52003228 National Natural Science Foundation of China (National Science Foundation of China)
- 52273197 National Natural Science Foundation of China (National Science Foundation of China)
- National Key Research and Development Program of China (2023YFB3810001), Shenzhen Key Laboratory of Functional Aggregate Materials (ZDSYS 20211021111400001), Science, Technology and Innovation Commission of Shenzhen Municipality (JCYJ 2021324134613038, KQTD 20210811090142053, JCYJ20220818103007014, GJHZ 20210705141810031), the Innovation and Technology Commission (ITC-CNERC14SC01), the Open Fund of Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates (2021-kllma-08), Guangzhou 510640, China (South China University of Technology). Guangzhou Science and Technology Planning Project (202201010439). Guangdong Basic and Applied Basic Research Foundation (2023A1515110346, 2021A1515110826). Guangzhou Science and Technology Planning Project (202201010439).
Collapse
Affiliation(s)
- Kun Zhou
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Lili Du
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Rui Ding
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Letian Xu
- Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou, China
| | - Shuai Shi
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Siyuan Wang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China
| | - Zaiyu Wang
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Guoqing Zhang
- University of Science and Technology of China, Hefei, Anhui, China
| | - Gang He
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| | - Zheng Zhao
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China.
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China.
| | - Ben Zhong Tang
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China.
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China.
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China.
- AIE Institute, Guangzhou Development District, Huangpu, Guangdong, China.
| |
Collapse
|
5
|
Cartagena J, Deshpande A, Rosenthal A, Tsang M, Hilal T, Rimsza L, Kurzrock R, Munoz J. Measurable Residual Disease in Mantle Cell Lymphoma: The Unbearable Lightness of Being Undetectable. Curr Oncol Rep 2024; 26:1664-1674. [PMID: 39641852 DOI: 10.1007/s11912-024-01620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE OF REVIEW This paper evaluates the benefits and limitations of detecting measurable residual disease (MRD) in mantle cell lymphoma (MCL) and assesses its prognostic value. It also aims to highlight the importance of detecting low MRD levels post-treatment and their application in clinical practice. RECENT FINDINGS Recent studies show that MRD levels predict relapse and survival outcomes in hematologic neoplasms, including MCL. RT-qPCR is currently the most used method due to its high reproducibility and sensitivity. Ideal MRD detection should be highly sensitive, cost-effective, and applicable to a wide demographic of patients. This paper concludes that MRD detection has prognostic value in MCL but faces limitations in sensitivity and specificity. Further research is needed to establish the significance of low MRD levels before integrating these methods into clinical practice. Improved MRD detection technologies and understanding their impact on clinical outcomes will guide better patient management in MCL.
Collapse
Affiliation(s)
- Julio Cartagena
- University of Puerto Rico School of Medicine, San Juan, PR, USA
| | | | - Allison Rosenthal
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Mazie Tsang
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Talal Hilal
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Lisa Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Razelle Kurzrock
- Michels Rare Cancers Research Laboratories, Froedtert and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Javier Munoz
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
6
|
Krieg S, Rohde T, Rausch T, Butthof L, Wendler-Link L, Eckert C, Breuhahn K, Galy B, Korbel J, Billmann M, Breinig M, Tschaharganeh DF. Mitoferrin2 is a synthetic lethal target for chromosome 8p deleted cancers. Genome Med 2024; 16:83. [PMID: 38886830 PMCID: PMC11181659 DOI: 10.1186/s13073-024-01357-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Somatic copy number alterations are a hallmark of cancer that offer unique opportunities for therapeutic exploitation. Here, we focused on the identification of specific vulnerabilities for tumors harboring chromosome 8p deletions. METHODS We developed and applied an integrative analysis of The Cancer Genome Atlas (TCGA), the Cancer Dependency Map (DepMap), and the Cancer Cell Line Encyclopedia to identify chromosome 8p-specific vulnerabilities. We employ orthogonal gene targeting strategies, both in vitro and in vivo, including short hairpin RNA-mediated gene knockdown and CRISPR/Cas9-mediated gene knockout to validate vulnerabilities. RESULTS We identified SLC25A28 (also known as MFRN2), as a specific vulnerability for tumors harboring chromosome 8p deletions. We demonstrate that vulnerability towards MFRN2 loss is dictated by the expression of its paralog, SLC25A37 (also known as MFRN1), which resides on chromosome 8p. In line with their function as mitochondrial iron transporters, MFRN1/2 paralog protein deficiency profoundly impaired mitochondrial respiration, induced global depletion of iron-sulfur cluster proteins, and resulted in DNA-damage and cell death. MFRN2 depletion in MFRN1-deficient tumors led to impaired growth and even tumor eradication in preclinical mouse xenograft experiments, highlighting its therapeutic potential. CONCLUSIONS Our data reveal MFRN2 as a therapeutic target of chromosome 8p deleted cancers and nominate MFNR1 as the complimentary biomarker for MFRN2-directed therapies.
Collapse
Affiliation(s)
- Stephan Krieg
- Helmholtz-University Group "Cell Plasticity and Epigenetic Remodeling", German Cancer Research Center (DKFZ), Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Rohde
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Tobias Rausch
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Luise Butthof
- Helmholtz-University Group "Cell Plasticity and Epigenetic Remodeling", German Cancer Research Center (DKFZ), Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lena Wendler-Link
- Helmholtz-University Group "Cell Plasticity and Epigenetic Remodeling", German Cancer Research Center (DKFZ), Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Eckert
- Helmholtz-University Group "Cell Plasticity and Epigenetic Remodeling", German Cancer Research Center (DKFZ), Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Bruno Galy
- Division of Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Korbel
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Maximilian Billmann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany.
| | - Marco Breinig
- Helmholtz-University Group "Cell Plasticity and Epigenetic Remodeling", German Cancer Research Center (DKFZ), Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.
| | - Darjus F Tschaharganeh
- Helmholtz-University Group "Cell Plasticity and Epigenetic Remodeling", German Cancer Research Center (DKFZ), Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
7
|
Shreenivas A, Nishizaki D, Lee S, Pabla S, Nesline M, Conroy JM, DePietro P, Kato S, Kurzrock R. Clinical and Biologic Correlates of ADORA2A Transcriptomic Expression in Cancer. Int J Mol Sci 2024; 25:4742. [PMID: 38731962 PMCID: PMC11083822 DOI: 10.3390/ijms25094742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
ADORA2A (adenosine A2a receptor) and ADORA2B propagate immunoregulatory signals, including restricting both innate and adaptive immunity, though recent data also suggest a tumor suppressor effect in certain settings. We evaluated the RNA expression from 514 tumors in a clinical-grade laboratory; 489 patients with advanced/metastatic disease had clinical outcome correlates. Transcript expression was standardized to internal housekeeping genes and ranked (0-100 scale) relative to 735 specimens from 35 different cancer types. Transcript abundance rank values were defined as "low/moderate" (0-74) or "high" (75-100) percentile RNA expression ranks. Overall, 20.8% of tumors had high ADORA2A (≥75 percentile RNA rank). The greatest proportion of high ADORA2A expressors was found in neuroendocrine and breast cancers and sarcomas, whereas the lowest was found in colorectal and ovarian cancers, albeit with patient-to-patient variability. In multivariable logistic regression analysis, there was a significant positive correlation between high ADORA2A RNA expression and a high expression of the immune checkpoint-related molecules PD-1 (p = 0.015), VISTA (p ≤ 0.001), CD38 (p = 0.031), and CD39 (p ≤ 0.001). In 217 immunotherapy-treated patients, high ADORA2A did not correlate significantly with progression-free (p = 0.51) or overall survival (OS) (p = 0.09) from the initiation of the checkpoint blockade. However, high versus not-high ADORA2A transcript expression correlated with longer OS from the time of advanced/metastatic disease (N = 489 patients; (HR 0.69 (95% CI 0.51-0.95) (p = 0.02)). Therefore, high ADORA2A transcript levels may be a favorable prognostic factor, unrelated to immunotherapy. Importantly, ascertaining co-expression patterns of ADORA2A with PD-1 and VISTA in individual tumors as a basis for the precision co-targeting of ADORA2A and these other checkpoint-related molecules warrants investigation in clinical trials.
Collapse
Affiliation(s)
- Aditya Shreenivas
- Department of Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA 92037, USA
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA 92037, USA
| | | | | | | | | | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA 92037, USA
| | - Razelle Kurzrock
- Department of Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA
- WIN Consortium, 24, rue Albert Thuret, 94550 Chevilly-Larue, France
- Department of Medical Oncology, University of Nebraska, Omaha, NE 68105, USA
| |
Collapse
|
8
|
Fujiwara Y, Kato S, Nishizaki D, Miyashita H, Lee S, Nesline MK, Conroy JM, DePietro P, Pabla S, Lippman SM, Kurzrock R. High indoleamine 2,3-dioxygenase transcript levels predict better outcome after front-line cancer immunotherapy. iScience 2024; 27:109632. [PMID: 38632994 PMCID: PMC11022045 DOI: 10.1016/j.isci.2024.109632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/07/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1), which catabolizes tryptophan, is a potential target to unlock the immunosuppressive tumor microenvironment. Correlations between IDO1 and immune checkpoint inhibitor (ICI) efficacy remain unclear. Herein, we investigated IDO1 transcript expression across cancers and clinical outcome correlations. High IDO1 transcripts were more frequent in uterine (54.2%) and ovarian cancer (37.2%) but varied between and within malignancies. High IDO1 RNA expression was associated with high expression of PD-L1 (immune checkpoint ligand), CXCL10 (an effector T cell recruitment chemokine), and STAT1 (a component of the JAK-STAT pathway) (all multivariable p < 0.05). PIK3CA and CTCF alterations were more frequent in the high IDO1 group. High IDO1 expression was an independent predictor of progression-free survival (adjusted HR = 0.44, 95% CI 0.20-0.99, p = 0.049) and overall survival (adjusted HR = 0.31, 95% CI 0.11-0.87, p = 0.026) after front-line ICIs. IDO1 expression warrants further exploration as a predictive biomarker for immunotherapy. Moreover, co-expressed immunoregulatory molecules merit exploration for co-targeting.
Collapse
Affiliation(s)
- Yu Fujiwara
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Beth Israel, New York, NY 10003, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA 92093, USA
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA 92093, USA
| | - Hirotaka Miyashita
- Division of Hematology and Oncology, Dartmouth Cancer Center. One Medical Center Drive, Lebanon, NH 03766, USA
| | - Suzanna Lee
- Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA 92093, USA
| | | | | | | | | | - Scott M. Lippman
- Center for Personalized Cancer Therapy, University of California, San Diego, Moores Cancer Center, La Jolla, CA 92093, USA
| | - Razelle Kurzrock
- MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
9
|
Moussa P, Kurzrock R, Nishizaki D, Miyashita H, Lee S, Nikanjam M, Pabla S, Nesline MK, Ko H, Conroy JM, DePietro P, Sicklick JK, Kato S. Transcriptomic analysis of GITR and GITR ligand reveals cancer immune heterogeneity with implications for GITR targeting. Am J Cancer Res 2024; 14:1634-1648. [PMID: 38726288 PMCID: PMC11076267 DOI: 10.62347/eced5481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/27/2024] [Indexed: 05/12/2024] Open
Abstract
Glucocorticoid-induced tumor necrosis factor related protein (GITR) is a transmembrane protein expressed mostly on CD25+CD4+ regulatory T-cells (Tregs) and upregulated on all T-cells upon activation. It is a T-cell co-stimulatory receptor and has demonstrated promising anti-tumor activity in pre-clinical studies. To date, however, the efficacy of GITR agonism has been discouraging in clinical trials. This study explores GITR and GITR ligand (GITR-L) ribonucleic acid (RNA) expression in solid tumors in an attempt to delineate causes for variable responses to GITR agonists. RNA expression levels of 514 patients with a variety of cancer types were normalized to internal housekeeping gene profiles and ranked as percentiles. 99/514 patients (19.3%) had high GITR expression (defined as ≥ 75th percentile). Breast and lung cancer had the highest proportion of patients with high GITR expression (39% and 35%, respectively). The expression of concomitant high GITR and low-moderate GITR-L expression (defined as <75th percentile) was present in 31% and 30% of patients with breast and lung cancer respectively. High GITR expression also showed a significant independent association with high RNA expression of other immune modulator proteins, namely, PD-L1 immunohistochemistry (IHC) ≥1 (odds ratio (OR) 2.15, P=0.008), CTLA4 (OR=2.17, P=0.05) and OX40 high RNA expression (OR=2.64, P=0.001). Overall, these results suggest that breast and lung cancer have a high proportion of patients with a GITR and GITR-L RNA expression profile that merits further investigation in GITR agonism studies. The association of high GITR expression with high CTLA4 and OX40 RNA expression, as well as positive PD-L1 IHC, provides a rationale for a combination approach targeting these specific immune modulator proteins in patients whose tumors show such co-expression.
Collapse
Affiliation(s)
- Peter Moussa
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology-Oncology, MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of WisconsinMilwaukee, WI, The United States
| | - Daisuke Nishizaki
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | - Hirotaka Miyashita
- Department of Medicine, Division of Hematology-Oncology, Dartmouth Cancer CenterLebanon, NH, The United States
| | - Suzanna Lee
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | - Mina Nikanjam
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | | | | | - Heidi Ko
- Labcorp OncologyDurham, NC, The United States
| | | | | | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, UC San DiegoSan Diego, CA, The United States
- Department of Pharmacology, UC San DiegoSan Diego, CA, The United States
- Structural and Functional Genomics Program, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| | - Shumei Kato
- Department of Medicine, Division of Hematology-Oncology, Moores Cancer Center, UC San DiegoLa Jolla, CA, The United States
| |
Collapse
|
10
|
Nishizaki D, Kurzrock R, Miyashita H, Adashek JJ, Lee S, Nikanjam M, Eskander RN, Patel H, Botta GP, Nesline MK, Pabla S, Conroy JM, DePietro P, Sicklick JK, Kato S. Viewing the immune checkpoint VISTA: landscape and outcomes across cancers. ESMO Open 2024; 9:102942. [PMID: 38503143 PMCID: PMC10966162 DOI: 10.1016/j.esmoop.2024.102942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Optimizing immune checkpoint inhibitor (ICI) therapy may require identification of co-targetable checkpoint pathways via immune profiling. Herein, we analyzed the transcriptomic expression and clinical correlates of V-domain immunoglobulin suppressor of T-cell activation (VISTA), a promising targetable checkpoint. PATIENTS AND METHODS RNA sequencing was carried out on 514 tissues reflecting diverse advanced/metastatic cancers. Expression of eight immune checkpoint markers [lymphocyte-activation gene 3 (LAG-3), tumor necrosis factor receptor superfamily 14 (TNFRSF14), programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), programmed death-ligand 2 (PD-L2), B- and T-lymphocyte attenuator (BTLA), T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), cytotoxic T-lymphocyte antigen 4 (CTLA-4)], in addition to VISTA, was analyzed, along with clinical outcomes. RESULTS High VISTA RNA expression was observed in 32% of tumors (66/514) and was the most common highly expressed checkpoint among the nine assessed. High VISTA expression was independently correlated with high BTLA, TIM-3, and TNFRSF14, and with a diagnosis of pancreatic, small intestine, and stomach cancer. VISTA transcript levels did not correlate with overall survival (OS) from metastatic/advanced disease in the pan-cancer cohort or with immunotherapy outcome (progression-free survival and OS from the start of ICI) in 217 ICI-treated patients. However, in ICI-treated pancreatic cancer patients (n = 16), median OS was significantly shorter (from immunotherapy initiation) for the high- versus not-high-VISTA groups (0.28 versus 1.21 years) (P = 0.047); in contrast, VISTA levels were not correlated with OS in 36 pancreatic cancer patients who did not receive ICI. CONCLUSION High VISTA expression correlates with high BTLA, TIM-3, and TNFRSF14 checkpoint-related molecules and with poorer post-immunotherapy survival in pancreatic cancer, consistent with prior literature indicating that VISTA is prominently expressed on CD68+ macrophages in pancreatic cancers and requiring validation in larger prospective studies. Immunomic analysis may be important for individualized precision immunotherapy.
Collapse
Affiliation(s)
- D Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla.
| | - R Kurzrock
- MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, USA; WIN Consortium, Paris, France
| | - H Miyashita
- Dartmouth Cancer Center, Hematology and Medical Oncology, Lebanon
| | - J J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore
| | - S Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - M Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - R N Eskander
- Center for Personalized Cancer Therapy and Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, Moores Cancer Center, La Jolla
| | - H Patel
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - G P Botta
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | | | | | | | | | - J K Sicklick
- Division of Surgical Oncology, Department of Surgery, Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, USA
| | - S Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla.
| |
Collapse
|
11
|
Adashek JJ, Kato S, Sicklick JK, Lippman SM, Kurzrock R. If it's a target, it's a pan-cancer target: Tissue is not the issue. Cancer Treat Rev 2024; 125:102721. [PMID: 38522181 PMCID: PMC11093268 DOI: 10.1016/j.ctrv.2024.102721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Cancer is traditionally diagnosed and treated on the basis of its organ of origin (e.g., lung or colon cancer). However, organ-of-origin diagnostics does not reveal the underlying oncogenic drivers. Fortunately, molecular diagnostics have advanced at a breathtaking pace, and it is increasingly apparent that cancer is a disease of the genome. Hence, we now have multiple genomic biomarker-based, tissue-agnostic Food and Drug Administration approvals for both gene- and immune-targeted therapies (larotrectinib/entrectinib, for NTRK fusions; selpercatinib, RET fusions; dabrafenib plus trametinib, BRAFV600E mutations; pembrolizumab/dostarlimab, microsatellite instability; and pembrolizumab for high tumor mutational burden; pemigatinib is also approved for FGFR1-rearranged myeloid/lymphoid neoplasms). There are emerging targets as well, including but not limited to ALK, BRCA and/or homologous repair deficiency, ERBB2 (HER2), IDH1/2, KIT, KRASG12C, NRG1, and VHL. Many tissue-agnostic approvals center on rare/ultra-rare biomarkers (often < 1 % of cancers), necessitating screening hundreds of tumors to find a single one harboring the cognate molecular alteration. Approval has generally been based on small single-arm studies (<30-100 patients) with high response rates (>30 % to > 75 %) of remarkable durability. Because of biomarker rarity, single-gene testing is not practical; next generation sequencing of hundreds of genes must be performed to obtain timely answers. Resistance to biomarker-driven therapeutics is often due to secondary mutations or co-driver gene defects; studies are now addressing the need for customized drug combinations matched to the complex molecular alteration portfolio in each tumor. Future investigation should expand tissue-agnostic therapeutics to encompass both hematologic and solid malignancies and include biomarkers beyond those that are DNA-based.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA; Department of Surgery, Division of Surgical Oncology, University of California San Diego, UC San Diego Health, San Diego, CA, USA; Department of Pharmacology, University of California San Diego, UC San Diego Health, San Diego, CA, USA
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee Wisconsin, USA; WIN Consortium, Paris France; University of Nebraska, United States.
| |
Collapse
|
12
|
Adashek JJ, Pandya C, Maragakis NJ, De P, Cohen PR, Kato S, Kurzrock R. Neuregulin-1 and ALS19 (ERBB4): at the crossroads of amyotrophic lateral sclerosis and cancer. BMC Med 2024; 22:74. [PMID: 38369520 PMCID: PMC10875826 DOI: 10.1186/s12916-024-03293-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/12/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Neuregulin-1 (NRG1) is implicated in both cancer and neurologic diseases such as amyotrophic lateral sclerosis (ALS); however, to date, there has been little cross-field discussion between neurology and oncology in regard to these genes and their functions. MAIN BODY Approximately 0.15-0.5% of cancers harbor NRG1 fusions that upregulate NRG1 activity and hence that of the cognate ERBB3/ERBB4 (HER3/HER4) receptors; abrogating this activity with small molecule inhibitors/antibodies shows preliminary tissue-agnostic anti-cancer activity. Notably, ERBB/HER pharmacologic suppression is devoid of neurologic toxicity. Even so, in ALS, attenuated ERBB4/HER4 receptor activity (due to loss-of-function germline mutations or other mechanisms in sporadic disease) is implicated; indeed, ERBB4/HER4 is designated ALS19. Further, secreted-type NRG1 isoforms may be upregulated (perhaps via a feedback loop) and could contribute to ALS pathogenesis through aberrant glial cell stimulation via enhanced activity of other (e.g., ERBB1-3/HER1-3) receptors and downstream pathways. Hence, pan-ERBB inhibitors, already in use for cancer, may be agents worthy of testing in ALS. CONCLUSION Common signaling cascades between cancer and ALS may represent novel therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Johns Hopkins Hospital, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Chinmayi Pandya
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | | | - Pradip De
- Cancer Genomics, Avera Cancer Institute, Sioux Falls, SD, USA
| | - Philip R Cohen
- Department of Dermatology, Davis Medical Center, University of California, Sacramento, CA, USA
- Touro University California College of Osteopathic Medicine, Vallejo, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- WIN Consortium, Paris, France.
- MCW Cancer Center, Milwaukee, WI, USA.
- University of Nebraska, Omaha, NE, USA.
| |
Collapse
|
13
|
Lim J, Kurzrock R, Nishizaki D, Miyashita H, Adashek JJ, Lee S, Pabla S, Nesline M, Conroy JM, DePietro P, Lippman SM, Kato S. Pan-cancer analysis of TIM-3 transcriptomic expression reveals high levels in pancreatic cancer and interpatient heterogeneity. Cancer Med 2024; 13:e6844. [PMID: 38132831 PMCID: PMC10807558 DOI: 10.1002/cam4.6844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), an immune checkpoint receptor, dampens immune function. TIM-3 antagonists have entered the clinic. METHODS We analyzed TIM-3 transcriptomic expression in 514 diverse cancers. Transcript abundance was normalized to internal housekeeping genes and ranked (0-100 percentile) to a reference population (735 tumors; 35 histologies [high≥75 percentile rank]). Ninety tumors (17.5%) demonstrated high TIM-3 expression. RESULTS TIM-3 expression varied between and within tumor types. However, high TIM-3 expression was more common in pancreatic cancer (20/55 tumors, 36.4%; odds ratio, 95% confidence interval (pancreatic vs. other tumors) = 3.176 (1.733-5.818; p < 0.001, multivariate]). High TIM-3 also significantly and independently correlated with high PD-L1 (p = 0.014) and high CTLA-4 (p < 0.001) transcriptomic expression (multivariate). CONCLUSIONS These observations indicate that TIM-3 RNA expression is heterogeneous, but more common in pancreatic cancer and in tumors exploiting PD-L1 and CTLA-4 checkpoints. Clinical trials with patient selection for matched immune-targeted combinations may be warranted.
Collapse
Affiliation(s)
| | - Razelle Kurzrock
- MCW Cancer CenterMilwaukeeWisconsinUSA
- WIN ConsortiumParisFrance
- University of NebraskaOmahaNebraskaUSA
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Hirotaka Miyashita
- Dartmouth Cancer Center, Hematology and Medical OncologyLebanonNew HampshireUSA
| | - Jacob J. Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer CenterThe Johns Hopkins HospitalBaltimoreMarylandUSA
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | | | | | | | | | - Scott M. Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| |
Collapse
|
14
|
Turner JH. Cancer Care by Committee to be Superseded by Personal Physician-Patient Partnership Informed by Artificial Intelligence. Cancer Biother Radiopharm 2023; 38:497-505. [PMID: 37366774 DOI: 10.1089/cbr.2023.0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Multidisciplinary tumor boards (MTBs) have become the reference standard of cancer management, founded upon randomized controlled trial (RCT) evidence-based guidelines. The inordinate delays inherent in awaiting formal regulatory agency approvals of novel therapeutic agents, and the rigidities and nongeneralizability of this regimented approach, often deny cancer patients timely access to effective innovative treatment. Reluctance of MTBs to accept theranostic care of patients with advanced neuroendocrine tumors (NETs) and metastatic castrate-resistant prostate cancer resulted in decades of delay in the incorporation of 177Lu-octreotate and 177Lu-prostate-specific membrane antigen (PSMA) into routine clinical oncology practice. Recent developments in immunotherapy and molecular targeted precision therapy, based on N-of-One individual multifactorial genome analyses, have greatly increased the complexity of decision-making. Burgeoning specialist workload and tight time frames now threaten to overwhelm the logistically, and emotionally, demanding MTB system. It is hypothesized that the advent of advanced artificial intelligence technology and Chatbot natural language algorithms will shift the cancer care paradigm from a MTB management model toward a personal physician-patient shared-care partnership for real-world practice of precision individualized holistic oncology.
Collapse
Affiliation(s)
- J Harvey Turner
- Department of Nuclear Medicine, Fiona Stanley Fremantle Hospitals Group, The University of Western Australia, Murdoch, Australia
| |
Collapse
|
15
|
Fountzilas E, Vo HH, Mueller P, Kurzrock R, Tsimberidou AM. Correlation between biomarkers and treatment outcomes in diverse cancers: a systematic review and meta-analysis of phase I and II immunotherapy clinical trials. Eur J Cancer 2023; 189:112927. [PMID: 37364526 PMCID: PMC10528229 DOI: 10.1016/j.ejca.2023.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Many immuno-oncology (IO) trials are conducted without biomarker selection. We performed a meta-analysis of phase I/II clinical trials evaluating immune checkpoint inhibitors (ICIs) to determine the association between biomarkers and clinical outcomes, if any. METHODS A PubMed search for phase I/II clinical trials with drugs approved by the Food and Drug Administration (labelled, off-label, combined with investigational ICIs or other treatment modalities) from 2018 to 2020 was performed. The objective response rate (ORR), progression-free survival (PFS) and overall survival (OS) were compared between biomarker-positive and biomarker-negative groups, using studies that explored the correlation of biomarkers with outcomes. RESULTS Overall, 174 clinical studies that included 19,178 patients were identified, and 132 studies investigated>30 correlative biomarkers that included PD-L1 expression (≥1%, 111 studies), tumour mutational burden (20 studies) and microsatellite instability/mismatch repair deficiency (10 studies). Overall, 123, 46 and 30 cohorts (drugs, tumour types or biomarkers) with 11,692, 3065, and 2256 patient outcomes for ORR, PFS and OS, respectively, were analysed in correlation with biomarkers. Meta-analyses demonstrated that ICIs in patients with biomarker-positive tumours were associated with higher ORR (odds ratio 2.15 [95% CI, 1.79-2.58], p < 0.0001); and longer PFS (hazard ratio [HR] 0.55 [95% CI, 0.45-0.67], p < 0.0001), and OS (HR 0.65 [95% CI, 0.53-0.80], p < 0.0001) compared with those with biomarker-negative tumours. Significance for ORR and PFS was retained in multivariate analysis (p < 0.001) (OS, not included owing to the small number of trials reporting OS). CONCLUSION Our data suggest that IO biomarkers should be used in patient selection for ICIs. Prospective studies are warranted.
Collapse
Affiliation(s)
- Elena Fountzilas
- Department of Medical Oncology, St Luke's Clinic, Thessaloniki, Greece; European University Cyprus, Limassol, Cyprus
| | - Henry Hiep Vo
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA
| | - Peter Mueller
- Department of Statistics and Data Science, The University of Texas at Austin, Austin, TX, USA
| | - Razelle Kurzrock
- WIN Consortium for Precision Medicine, Paris, France; Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Apostolia-Maria Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA.
| |
Collapse
|
16
|
Moeller A, Kurzrock R, Botta GP, Adashek JJ, Patel H, Lee S, Pabla S, Nesline MK, Conroy J, Sicklick JK, Kato S. Challenges and prospects of CSF1R targeting for advanced malignancies. Am J Cancer Res 2023; 13:3257-3265. [PMID: 37560003 PMCID: PMC10408490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/28/2023] [Indexed: 08/11/2023] Open
Abstract
CSF1R expression modulates tumor-associated macrophages, making CSF1R blockade an appealing immune-modulating therapeutic target. We evaluated the correlation between CSF1R tumor RNA expression and outcome (pan-cancer setting). RNA expression was ranked as a percentile (0-100) using a standardized internal reference population (735 tumors; 35 histologies). Among 514 patients, there was no difference in survival from biopsy between high and low CSF1R expressors (< 50 percentile versus ≥ 50 percentile rank). There was also no significant difference in median progression-free or overall survival (from treatment) based on CSF1R expression in 21 patients who received CSF1R inhibitors (all p values ≥ 0.08). Concurrent upregulation of ≥ 2 additional immune checkpoint markers (e.g. PD-L1, BTLA, CTLA4, LAG3, TIM3) was observed in all tumor samples with CSF1R expression ≥ 50th percentile. Pending further large prospective studies, patients with high tumor CSF1R expression may need treatment that co-targets the specific immune checkpoint pathways activated in order to impact outcome.
Collapse
Affiliation(s)
- Ann Moeller
- Sharp Rees-Stealy Medical GroupSan Diego, CA, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center and Genomic Sciences and Precision Medicine CenterMilwaukee, WI, USA
- Worldwide Innovative Network (WIN) for Personalized Cancer TherapyParis, France
| | - Gregory P Botta
- Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa Jolla, CA, USA
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins HospitalBaltimore, MD, USA
| | - Hitendra Patel
- Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa Jolla, CA, USA
| | - Suzanna Lee
- Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa Jolla, CA, USA
| | | | | | | | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, UC San Diego School of MedicineSan Diego, CA, USA
| | - Shumei Kato
- Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa Jolla, CA, USA
| |
Collapse
|
17
|
Tateo V, Marchese PV, Mollica V, Massari F, Kurzrock R, Adashek JJ. Agnostic Approvals in Oncology: Getting the Right Drug to the Right Patient with the Right Genomics. Pharmaceuticals (Basel) 2023; 16:ph16040614. [PMID: 37111371 PMCID: PMC10144220 DOI: 10.3390/ph16040614] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background: The oncology field has drastically changed with the advent of precision medicine, led by the discovery of druggable genes or immune targets assessed through next-generation sequencing. Biomarker-based treatments are increasingly emerging, and currently, six tissue-agnostic therapies are FDA-approved. (2) Methods: We performed a review of the literature and reported the trials that led to the approval of tissue-agnostic treatments and ongoing clinical trials currently investigating novel biomarker-based approaches. (3) Results: We discussed the approval of agnostic treatments: pembrolizumab and dostarlimab for MMRd/MSI-H, pembrolizumab for TMB-H, larotrectinib and entrectinib for NTRK-fusions, dabrafenib plus trametinib for BRAF V600E mutation, and selpercatinib for RET fusions. In addition, we reported novel clinical trials of biomarker-based approaches, including ALK, HER2, FGFR, and NRG1. (4) Conclusions: Precision medicine is constantly evolving, and with the improvement of diagnostic tools that allow a wider genomic definition of the tumor, tissue-agnostic targeted therapies are a promising treatment strategy tailored to the specific tumor genomic profile, leading to improved survival outcomes.
Collapse
Affiliation(s)
- Valentina Tateo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Paola Valeria Marchese
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40127 Bologna, Italy
| | - Razelle Kurzrock
- MCW Cancer Center, Milwaukee, WI 53226, USA
- WIN Consortium, San Diego, CA 92093, USA
- Department of Oncology, University of Nebraska, Omaha, NE 68198, USA
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
18
|
Adashek JJ, Sapkota S, de Castro Luna R, Seiwert TY. Complete response to alectinib in ALK-fusion metastatic salivary ductal carcinoma. NPJ Precis Oncol 2023; 7:36. [PMID: 37041305 PMCID: PMC10090142 DOI: 10.1038/s41698-023-00378-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/30/2023] [Indexed: 04/13/2023] Open
Abstract
The advent of next-generation sequencing (NGS) has allowed for the identification of novel therapeutic targets for patients with uncommon cancers. It is well known that fusion translocations are potent driver of cancer pathogenesis and can render tumors exquisitely sensitive to matching targeted therapies. Here we describe a patient with ALK-fusion positive widely metastatic salivary ductal carcinoma, who achieved a durable complete response from alectinib, a potent and specific ALK tyrosine kinase inhibitor. This case serves as another reminder that ALK-fusions can be targeted regardless of histology and can afford patients dramatic and durable benefit. It also emphasizes the need for insurance coverage for such beneficial therapies. While ALK fusions are exceedingly rare in salivary ductal carcinoma, the presence of multiple other targetable aberrations supports the recommendation for universal NGS testing for such tumors.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Surendra Sapkota
- Department of Internal Medicine, Saint Agnes Hospital, Baltimore, MD, USA
| | - Rodrigo de Castro Luna
- Department of Radiology and Radiological Science, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Tanguy Y Seiwert
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| |
Collapse
|
19
|
Mangiante L, Alcala N, Sexton-Oates A, Di Genova A, Gonzalez-Perez A, Khandekar A, Bergstrom EN, Kim J, Liu X, Blazquez-Encinas R, Giacobi C, Le Stang N, Boyault S, Cuenin C, Tabone-Eglinger S, Damiola F, Voegele C, Ardin M, Michallet MC, Soudade L, Delhomme TM, Poret A, Brevet M, Copin MC, Giusiano-Courcambeck S, Damotte D, Girard C, Hofman V, Hofman P, Mouroux J, Cohen C, Lacomme S, Mazieres J, de Montpreville VT, Perrin C, Planchard G, Rousseau N, Rouquette I, Sagan C, Scherpereel A, Thivolet F, Vignaud JM, Jean D, Ilg AGS, Olaso R, Meyer V, Boland-Auge A, Deleuze JF, Altmuller J, Nuernberg P, Ibáñez-Costa A, Castaño JP, Lantuejoul S, Ghantous A, Maussion C, Courtiol P, Hernandez-Vargas H, Caux C, Girard N, Lopez-Bigas N, Alexandrov LB, Galateau-Salle F, Foll M, Fernandez-Cuesta L. Multiomic analysis of malignant pleural mesothelioma identifies molecular axes and specialized tumor profiles driving intertumor heterogeneity. Nat Genet 2023; 55:607-618. [PMID: 36928603 PMCID: PMC10101853 DOI: 10.1038/s41588-023-01321-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 01/26/2023] [Indexed: 03/17/2023]
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with rising incidence and challenging clinical management. Through a large series of whole-genome sequencing data, integrated with transcriptomic and epigenomic data using multiomics factor analysis, we demonstrate that the current World Health Organization classification only accounts for up to 10% of interpatient molecular differences. Instead, the MESOMICS project paves the way for a morphomolecular classification of MPM based on four dimensions: ploidy, tumor cell morphology, adaptive immune response and CpG island methylator profile. We show that these four dimensions are complementary, capture major interpatient molecular differences and are delimited by extreme phenotypes that-in the case of the interdependent tumor cell morphology and adapted immune response-reflect tumor specialization. These findings unearth the interplay between MPM functional biology and its genomic history, and provide insights into the variations observed in the clinical behavior of patients with MPM.
Collapse
Affiliation(s)
- Lise Mangiante
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Nicolas Alcala
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
| | - Alexandra Sexton-Oates
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
| | - Alex Di Genova
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
- Instituto de Ciencias de la Ingeniería, Universidad de O'Higgins, Rancagua, Chile
- Centro de Modelamiento Matemático UMI-CNRS 2807, Universidad de Chile, Santiago, Chile
| | - Abel Gonzalez-Perez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Azhar Khandekar
- Department of Cellular and Molecular Medicine, Department of Bioengineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Erik N Bergstrom
- Department of Cellular and Molecular Medicine, Department of Bioengineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jaehee Kim
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Xiran Liu
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Ricardo Blazquez-Encinas
- Maimonides Biomedical Research Institute of Cordoba, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Colin Giacobi
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
| | - Nolwenn Le Stang
- UMR INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, MESOPATH-MESOBANK, Department of Biopathology, Cancer Centre Léon Bérard, Lyon, France
| | - Sandrine Boyault
- Cancer Genomic Platform, Translational Research and Innovation Department, Centre Léon Bérard, Lyon, France
| | - Cyrille Cuenin
- EpiGenomics and Mechanisms Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
| | - Severine Tabone-Eglinger
- UMR INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, MESOPATH-MESOBANK, Department of Biopathology, Cancer Centre Léon Bérard, Lyon, France
| | - Francesca Damiola
- UMR INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, MESOPATH-MESOBANK, Department of Biopathology, Cancer Centre Léon Bérard, Lyon, France
| | - Catherine Voegele
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
| | - Maude Ardin
- Tumor Escape, Resistance and Immunity Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Marie-Cecile Michallet
- Tumor Escape, Resistance and Immunity Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Lorraine Soudade
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
| | - Tiffany M Delhomme
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Arnaud Poret
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
| | | | - Marie-Christine Copin
- University of Lille, Centre Hospitalier Universitaire Lille, Institut de Pathologie, Tumorothèque du Centre de Référence Régional en Cancérologie, Lille, France
| | | | - Diane Damotte
- Centre de Recherche des Cordeliers, Inflammation, Complement and Cancer Team, Sorbonne Université, INSERM, Université de Paris, Paris, France
- Department of Pathology, Hôpitaux Universitaire Paris Centre, Tumorothèque/CRB Cancer, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Cecile Girard
- Tumorothèque Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Veronique Hofman
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Nice Center Hospital, FHU OncoAge, Biobank BB-0033-00025 and IRCAN Inserm U1081/CNRS 7284, Nice, France
| | - Paul Hofman
- Université Côte d'Azur, Laboratory of Clinical and Experimental Pathology, Nice Center Hospital, FHU OncoAge, Biobank BB-0033-00025 and IRCAN Inserm U1081/CNRS 7284, Nice, France
| | - Jérôme Mouroux
- Université Côte d'Azur, Department of Thoracic Surgery, Nice Center Hospital, FHU OncoAge and IRCAN Inserm U1081/CNRS 7284, Nice, France
| | - Charlotte Cohen
- Department of Thoracic Surgery, FHU OncoAge, Nice Pasteur Hospital, Université Côte d'Azur, Nice, France
| | - Stephanie Lacomme
- Nancy Regional University Hospital, Centre Hospitalier Régional Universitaire, CRB BB-0033-00035, INSERM U1256, Nancy, France
| | - Julien Mazieres
- Toulouse University Hospital, Université Paul Sabatier, Toulouse, France
| | | | - Corinne Perrin
- Hospices Civils de Lyon, Institut de Pathologie, Centre de Ressources Biologiques des HCL, Tissu-Tumorothèque Est, Lyon, France
| | - Gaetane Planchard
- Centre Hospitalier Universitaire de Caen, MESOPATH Regional Center, Caen, France
| | - Nathalie Rousseau
- Centre Hospitalier Universitaire de Caen, MESOPATH Regional Center, Caen, France
| | - Isabelle Rouquette
- Centre de Pathologie des Côteaux, Centre de Ressources Biologiques (CRB Cancer), IUCT Oncopole, Toulouse, France
| | - Christine Sagan
- Tumorothèque Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Arnaud Scherpereel
- University of Lille, Centre Hospitalier Universitaire Lille, INSERM, OncoThAI, NETMESO Network, Lille, France
| | - Francoise Thivolet
- Hospices Civils de Lyon, Institut de Pathologie, Centre de Ressources Biologiques des HCL, Tissu-Tumorothèque Est, Lyon, France
| | - Jean-Michel Vignaud
- Department of Biopathology, Centre Hospitalier Régional Universitaire de Nancy, Vandoeuvre-les-Nancy, France
- BRC, BB-0033-00035, Centre Hospitalier Régional Universitaire de Nancy, Vandoeuvre-les-Nancy, France
| | - Didier Jean
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors, Paris, France
| | | | - Robert Olaso
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, Evry, France
| | - Vincent Meyer
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, Evry, France
| | - Anne Boland-Auge
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, Evry, France
| | - Jean-Francois Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, Evry, France
| | | | | | - Alejandro Ibáñez-Costa
- Maimonides Biomedical Research Institute of Cordoba, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Sylvie Lantuejoul
- UMR INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, MESOPATH-MESOBANK, Department of Biopathology, Cancer Centre Léon Bérard, Lyon, France
- Grenoble Alpes University, Saint-Martin-d'Hères, France
| | - Akram Ghantous
- EpiGenomics and Mechanisms Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France
| | | | | | - Hector Hernandez-Vargas
- UMR INSERM 1052, CNRS 5286, UCBL1, Centre Léon Bérard, Lyon, France
- Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Christophe Caux
- Tumor Escape, Resistance and Immunity Department, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Nicolas Girard
- Institut Curie, Institut du Thorax Curie Montsouris, Paris, France
- Université de Versailles Saint-Quentin-en-Yvelines, Université Paris-Saclay, Versailles, France
| | - Nuria Lopez-Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Cáncer, Instituto de Salud Carlos III, Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, Department of Bioengineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Françoise Galateau-Salle
- UMR INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, MESOPATH-MESOBANK, Department of Biopathology, Cancer Centre Léon Bérard, Lyon, France
| | - Matthieu Foll
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France.
| | - Lynnette Fernandez-Cuesta
- Rare Cancers Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer/World Health Organization, Lyon, France.
| |
Collapse
|
20
|
Gupta A, Kurzrock R, Adashek JJ. Evolution of the Targeted Therapy Landscape for Cholangiocarcinoma: Is Cholangiocarcinoma the 'NSCLC' of GI Oncology? Cancers (Basel) 2023; 15:1578. [PMID: 36900367 PMCID: PMC10000383 DOI: 10.3390/cancers15051578] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023] Open
Abstract
In the past two decades, molecular targeted therapy has revolutionized the treatment landscape of several malignancies. Lethal malignancies such as non-small cell lung cancer (NSCLC) have become a model for precision-matched immune- and gene-targeted therapies. Multiple small subgroups of NSCLC defined by their genomic aberrations are now recognized; remarkably, taken together, almost 70% of NSCLCs now have a druggable anomaly. Cholangiocarcinoma (CCA) is a rare tumor with a poor prognosis. Novel molecular alterations have been recently identified in patients with CCA, and the potential for targeted therapy is being realized. In 2019, a fibroblast growth factor receptor 2 (FGFR2) inhibitor, pemigatinib, was the first approved targeted therapy for patients with locally advanced or metastatic intrahepatic CCA who had FGFR2 gene fusions or rearrangement. More regulatory approvals for matched targeted therapies as second-line or subsequent treatments in advanced CCA followed, including additional drugs that target FGFR2 gene fusion/rearrangement. Recent tumor-agnostic approvals include (but are not limited to) drugs that target mutations/rearrangements in the following genes and are hence applicable to CCA: isocitrate dehydrogenase 1 (IDH1); neurotrophic tropomyosin-receptor kinase (NTRK); the V600E mutation of the BRAF gene (BRAFV600E); and high tumor mutational burden, high microsatellite instability, and gene mismatch repair-deficient (TMB-H/MSI-H/dMMR) tumors. Ongoing trials investigate HER2, RET, and non-BRAFV600E mutations in CCA and improvements in the efficacy and safety of new targeted treatments. This review aims to present the current status of molecularly matched targeted therapy for advanced CCA.
Collapse
Affiliation(s)
- Amol Gupta
- Department of Medicine, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Razelle Kurzrock
- WIN Consortium, San Diego, CA 92093, USA
- Division of Hematology and Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA
- Division of Hematology and Oncology, University of Nebraska, Omaha, NE 68182, USA
| | - Jacob J. Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
21
|
Shaya J, Kato S, Adashek JJ, Patel H, Fanta PT, Botta GP, Sicklick JK, Kurzrock R. Personalized matched targeted therapy in advanced pancreatic cancer: a pilot cohort analysis. NPJ Genom Med 2023; 8:1. [PMID: 36670111 PMCID: PMC9860045 DOI: 10.1038/s41525-022-00346-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/07/2022] [Indexed: 01/21/2023] Open
Abstract
Despite progress, 2-year pancreatic cancer survival remains dismal. We evaluated a biomarker-driven, combination/N-of-one strategy in 18 patients (advanced/metastatic pancreatic cancer) (from Molecular Tumor Board). Targeted agents administered/patient = 2.5 (median) (range, 1-4); first-line therapy (N = 5); second line, (N = 13). Comparing patients (high versus low degrees of matching) (matching score ≥50% versus <50%; reflecting number of alterations matched to targeted agents divided by number of pathogenic alterations), survival was significantly longer (hazard ratio [HR] 0.24 (95% confidence interval [CI], 0.078-0.76, P = 0.016); clinical benefit rates (CBR) (stable disease ≥6 months/partial/complete response) trended higher (45.5 vs 0.0%, P = 0.10); progression-free survival, HR, 95% CI, 0.36 (0.12-1.10) (p = 0.075). First versus ≥2nd-line therapy had higher CBRs (80.0 vs 7.7%, P = 0.008). No grade 3-4 toxicities occurred. The longest responder achieved partial remission (17.5 months) by co-targeting MEK and CDK4/6 alterations (chemotherapy-free). Therefore, genomically matched targeted agent combinations were active in these advanced pancreatic cancers. Larger prospective trials are warranted.
Collapse
Affiliation(s)
- Justin Shaya
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA.
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, La Jolla, CA, USA.
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Hitendra Patel
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Paul T Fanta
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Gregory P Botta
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at The Johns Hopkins Hospital, Baltimore, MD, USA
- Department of Surgery, Division of Surgical Oncology, University of California San Diego, UC San Diego Health, San Diego, CA, USA
- Department of Pharmacology, University of California San Diego, UC San Diego Health, San Diego, CA, USA
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA
- WIN Consortium, Paris, France
- University of Nebraska, Lincoln, NE, USA
| |
Collapse
|
22
|
Adashek JJ, Subbiah V, Westphalen CB, Naing A, Kato S, Kurzrock R. Cancer: slaying the nine-headed Hydra. Ann Oncol 2023; 34:61-69. [PMID: 35931318 PMCID: PMC10923524 DOI: 10.1016/j.annonc.2022.07.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 02/03/2023] Open
Abstract
Modern medicine continues to evolve, and the treatment armamentarium for various diseases grows more individualized across a breadth of medical disciplines. Cure rates for infectious diseases that were previously pan-fatal approach 100% because of the identification of the specific pathogen(s) involved and the use of appropriate combinations of drugs, where needed, to completely extinguish infection and hence prevent emergence of resistant strains. Similarly, with the assistance of technologies such as next-generation sequencing and immunomic analysis as part of the contemporary oncology armory, therapies can be tailored to each tumor. Importantly, molecular interrogation has revealed that metastatic cancers are distinct from each other and complex. Therefore, it is conceivable that rational personalized drug combinations will be needed to eradicate cancers, and eradication will be necessary to mitigate clonal evolution and resistance.
Collapse
Affiliation(s)
- J J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore.
| | - V Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - C B Westphalen
- Comprehensive Cancer Center Munich and Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - A Naing
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California, San Diego
| | - R Kurzrock
- WIN Consortium, San Diego; MCW Cancer Center, Milwaukee; University of Nebraska, Omaha, USA.
| |
Collapse
|
23
|
Wahida A, Buschhorn L, Fröhling S, Jost PJ, Schneeweiss A, Lichter P, Kurzrock R. The coming decade in precision oncology: six riddles. Nat Rev Cancer 2023; 23:43-54. [PMID: 36434139 DOI: 10.1038/s41568-022-00529-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 11/27/2022]
Abstract
High-throughput methods to investigate tumour omic landscapes have quickly catapulted cancer specialists into the precision oncology era. The singular lesson of precision oncology might be that, for it to be precise, treatment must be personalized, as each cancer's complex molecular and immune landscape differs from patient to patient. Transformative therapies include those that are targeted at the sequelae of molecular abnormalities or at immune mechanisms, and, increasingly, pathways previously thought to be undruggable have become druggable. Critical to applying precision medicine is the concept that the right combination of drugs must be chosen for each patient and used at the right stage of the disease. Multiple puzzles remain that complicate therapy choice, including evidence that deleterious mutations are common in normal tissues and non-malignant conditions. The host's role is also likely to be key in determining treatment response, especially for immunotherapy. Indeed, maximizing the impact of immunotherapy will require omic analyses to match the right immune-targeted drugs to the individualized patient and tumour setting. In this Perspective, we discuss six key riddles that must be solved to optimize the application of precision oncology to otherwise lethal malignancies.
Collapse
Affiliation(s)
- Adam Wahida
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany.
- Medical Department III for Hematology and Oncology, Klinikum rechts der Isar, TUM School of Medicine, Technical University of Munich, Munich, Germany.
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.
| | - Lars Buschhorn
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.
| | - Stefan Fröhling
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Philipp J Jost
- Division of Clinical Oncology, Department of Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Schneeweiss
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Razelle Kurzrock
- WIN Consortium, Paris, France.
- Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
24
|
Fujiwara Y, Kato S, Nesline MK, Conroy JM, DePietro P, Pabla S, Kurzrock R. Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy. Cancer Treat Rev 2022; 110:102461. [PMID: 36058143 DOI: 10.1016/j.ctrv.2022.102461] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/18/2022] [Accepted: 08/26/2022] [Indexed: 11/02/2022]
Abstract
Strategies for unlocking immunosuppression in the tumor microenvironment have been investigated to overcome resistance to first-generation immune checkpoint blockade with anti- programmed cell death protein 1 (PD-1)/ programmed death-ligand 1 (PD-L1) and anti-cytotoxic T-lymphocyte associated protein 4 (CTLA-4) agents. Indoleamine 2,3-dioxygenase (IDO) 1, an enzyme catabolizing tryptophan to kynurenine, creates an immunosuppressive environment in preclinical studies. Early phase clinical trials investigating inhibition of IDO1, especially together with checkpoint blockade, provided promising results. Unfortunately, the phase 3 trial of the IDO1 inhibitor epacadostat combined with the PD-1 inhibitor pembrolizumab did not show clinical benefit when compared with pembrolizumab monotherapy in patients with advanced malignant melanoma, which dampened enthusiasm for IDO inhibitors. Even so, several molecules, such as the aryl hydrocarbon receptor and tryptophan 2,3-dioxygenase, were reported as additional potential targets for the modulation of the tryptophan pathway, which might enhance clinical effectiveness. Furthermore, the combination of IDO pathway blockade with agents inhibiting other signals, such as those generated by PIK3CA mutations that may accompany IDO1 upregulation, may be a novel way to enhance activity. Importantly, IDO1 expression level varies by tumor type and among patients with the same tumor type, suggesting that patient selection based on expression levels of IDO1 may be warranted in clinical trials.
Collapse
Affiliation(s)
- Yu Fujiwara
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Beth Israel, New York, NY, United States.
| | - Shumei Kato
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, United States.
| | | | | | | | | | - Razelle Kurzrock
- MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
25
|
Cohen PR, Kurzrock R. Dermatologic Disease-Directed Targeted Therapy (D 3T 2): The Application of Biomarker-Based Precision Medicine for the Personalized Treatment of Skin Conditions-Precision Dermatology. Dermatol Ther (Heidelb) 2022; 12:2249-2271. [PMID: 36121579 PMCID: PMC9515268 DOI: 10.1007/s13555-022-00801-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/23/2022] [Indexed: 11/03/2022] Open
Abstract
Precision dermatology uses individualized dermatologic disease-directed targeted therapy (D3T2) for the management of dermatoses and for the evaluation and therapy of cutaneous malignancies. Personalized/precision strategies are based on biomarkers that are most frequently derived from tissue transcriptomic expression or genomic sequencing or from circulating cytokines. For instance, the pathologic diagnosis of a pigmented lesion and determining the prognosis of a malignant melanocytic neoplasm can be enhanced by genomic/transcriptomic analysis. In addition to biopsy, innovative techniques have been developed for obtaining transcriptomes in skin conditions; as an example, patches can be applied to a psoriasis plaque for a few minutes to capture the epidermis/upper dermis transcriptome. Atopic dermatitis and prurigo nodularis may also be candidate conditions for precision dermatology. Precision dermatology has a role in managing melanoma and nonmelanoma skin cancers and rare cutaneous tumors-such as perivascular epithelioid cell tumor (PEComa)-that can originate in or metastasize to the skin. For instance, advanced/metastatic basal cell carcinomas can be treated with Hedgehog inhibitors (vismodegib and sonidegib) targeting the smoothened (SMO) or patched 1 (PTCH1) gene alterations that are a hallmark of these cancers and activate the Hedgehog pathway. Advanced/metastatic basal and cutaneous squamous cell cancers often have a high tumor mutational burden (which predicts immunotherapy response); immune checkpoint blockade with cemiplimab, a programmed cell death protein 1 (PD1) inhibitor, is now approved for these malignancies. Gene expression profiling of primary cutaneous squamous cell carcinoma can identify those individuals at high risk for subsequent metastases. In the realm of rare neoplasms, PEComas-which can originate in the skin, albeit uncommonly-have tuberous sclerosis complex 1 (TSC1)/tuberous sclerosis complex 2 (TSC2) gene alterations, which activate mammalian target of rapamycin (mTOR) signaling, and can be suppressed by nab-sirolimus, now approved for this condition. In summary, precision dermatologic techniques/strategies are an important emerging approach for evaluation and management of skin disorders and cutaneous neoplasms, and may serve as a paradigm for the application of precision medicine beyond dermatology.
Collapse
Affiliation(s)
- Philip R Cohen
- Department of Dermatology, Davis Medical Center, University of California, Sacramento, CA, USA.
- Touro University California College of Osteopathic Medicine, Vallejo, CA, USA.
- University of California, 10991 Twinleaf Court, San Diego, CA, 92131, USA.
| | - Razelle Kurzrock
- Department of Medicine, Medical College of Wisconsin Cancer Center and Genome Sciences and Precision Medicine Center, Milwaukee, WI, USA
- Worldwide Innovative Network (WIN) for Personalized Cancer Therapy, Villejuif, France
| |
Collapse
|
26
|
Mollica V, Massari F, Rizzo A, Ferrara R, Menta AK, Adashek JJ. Genomics and Immunomics in the Treatment of Urothelial Carcinoma. Curr Oncol 2022; 29:3499-3518. [PMID: 35621673 PMCID: PMC9139747 DOI: 10.3390/curroncol29050283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 12/21/2022] Open
Abstract
Urothelial carcinoma is a complex cancer with genomic immunomic drivers that have prognostic and predictive treatment implications. Identifying potential targetable alterations via next-generation sequencing and RNA sequencing may allow for elucidation of such targets and exploitation with targeted therapeutics. The role of immunotherapy in treating urothelial carcinoma has shown benefit, but it is unclear in which patients immunotherapeutics have the highest yield. Continuing efforts into better identifying which patients may benefit most from targeted therapies, immunotherapies, and combination therapies may ultimately lead to improved outcomes for patients with this disease.
Collapse
Affiliation(s)
- Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138 Bologna, Italy; (V.M.); (F.M.)
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138 Bologna, Italy; (V.M.); (F.M.)
| | - Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico ‘Don Tonino Bello’, I.R.C.C.S. Istituto Tumori ‘Giovanni Paolo II’, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Roberto Ferrara
- Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Istituto Nazionale dei Tumori, 20133 Milan, Italy;
- Molecular Immunology Unit, Department of Research, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Arjun K. Menta
- Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Jacob J. Adashek
- Department of Internal Medicine, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
27
|
Adashek JJ, Breunig JJ, Posadas E, Bhowmick NA, Ellis L, Freedland SJ, Kim H, Figlin R, Gong J. First-line Immune Checkpoint Inhibitor Combinations in Metastatic Renal Cell Carcinoma: Where Are We Going, Where Have We Been? Drugs 2022; 82:439-453. [PMID: 35175588 DOI: 10.1007/s40265-022-01683-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2022] [Indexed: 01/03/2023]
Abstract
The combination of targeted therapy and immunotherapy in the treatment of metastatic renal cell carcinoma (mRCC) has significantly improved outcomes for many patients. There are multiple FDA-approved regimens for the frontline setting based on numerous randomized Phase III trials. Despite these efforts, there remains a conundrum of identifying a biomarker-driven approach for these patients and it is unclear how to predict which patients are most likely to respond to these agents. This is due, in part, to an incomplete understanding of how these drug combinations work. The use of tyrosine kinase inhibitors that have multiple 'off-target' effects may lend themselves to the benefits observed when given in combination with immunotherapy. Further, targeting multiple clones within a patient's heterogenic tumor that are responsive to targeted therapy and others that are responsive to immunotherapy may also explain some level of improved response rates to the combination approaches compared to monotherapies. This review highlights the 5 FDA-approved regimens for mRCC in the frontline setting and offers insights into potential mechanisms for improved outcomes seen in these combination approaches.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Internal Medicine, University of South Florida, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Joshua J Breunig
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Edwin Posadas
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Neil A Bhowmick
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Leigh Ellis
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Stephen J Freedland
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA.,Division of Urology, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Section of Urology, Durham VA Medical Center, Durham, NC, USA
| | - Hyung Kim
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA.,Division of Urology, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robert Figlin
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Jun Gong
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA.
| |
Collapse
|