251
|
Khandpur U, Haile B, Makary MS. Early-Stage Renal Cell Carcinoma Locoregional Therapies: Current Approaches and Future Directions. Clin Med Insights Oncol 2024; 18:11795549241285390. [PMID: 39435052 PMCID: PMC11492234 DOI: 10.1177/11795549241285390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 09/03/2024] [Indexed: 10/23/2024] Open
Abstract
Renal cell carcinoma (RCC) is the most common primary renal malignancy. Prevalence of RCC in developed countries has slowly increased. Although partial or total nephrectomy has been the first-line treatment for early-stage RCC, improved or similar safety and treatment outcomes with locoregional therapies have challenged this paradigm. In this review, we explore locoregional techniques for early-stage RCC, including radiofrequency ablation, cryoablation, and microwave ablation with a focus on procedural technique, patient selection, and safety/treatment outcomes. Furthermore, we discuss future advances and novel techniques, including radiomics, combination therapy, high-intensity focused ultrasound, and catheter-directed techniques.
Collapse
Affiliation(s)
- Umang Khandpur
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bereket Haile
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mina S Makary
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
252
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
253
|
Majidova N, Seyyar M, Bayraktar DI, Dinç G, İsgandarov E, Huseynov J, Yaşar A, Çelebi A, Sever N, Kocaaslan E, Erel P, Ağyol Y, Güren AK, Arıkan R, Işık S, Ercelep Ö, Demirağ G, Kefeli U, Köstek O, Bayoğlu İV, Sarı M. Which factors help to determine the long-term response to first-line tyrosine kinase inhibitors in patients with metastatic renal cell carcinoma: A Turkish multi-centre study. BIOMOLECULES & BIOMEDICINE 2024; 24:1776-1784. [PMID: 38920621 PMCID: PMC11496851 DOI: 10.17305/bb.2024.10512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024]
Abstract
Many developing countries lack access to recommended first-line treatments for metastatic renal cell carcinoma (mRCC), such as immune checkpoint inhibitors (ICIs) or ICI-tyrosine kinase inhibitor (TKI) combinations. As a result, predictive markers are necessary to identify patients who may benefit from single-agent TKIs for long-term response. This study aims to identify such parameters. This was a multi-centre, retrospective study of patients with mRCC who were undergoing first-line treatment with sunitinib or pazopanib. Patients who had been diagnosed with mRCC and had not experienced disease progression for 36 months or more were deemed to have achieved a long-term response. Predictive clinical and pathological characteristics of patients who did not experience long-term disease progression were investigated. A total of 320 patients from four hospitals were included in the study. The median age of the patients was 60 years (range 20-89 years). According to the International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) risk classification, 109 patients were classified as having favourable risk and 211 were in the intermediate-poor risk group. The median progression-free survival (PFS) and overall survival (OS) for all patients were 12.5 months and 76.4 months, respectively. In the long-term responder's group, the median PFS was 78.4 months. Among all patients, prior nephrectomy, the Eastern Cooperative Oncology Group (ECOG) Performance Status (PS) <1, and the absence of brain metastasis were predictive factors for long-term response. For patients in the favourable risk group, the lack of brain metastasis was a predictor of long-term response. In the intermediate-poor risk group, prior nephrectomy and ECOG PS <1 were predictive factors for long-term response. Some individuals with mRCC may experience a durable response to TKIs. The likelihood of a long-term response can be determined by factors such as nephrectomy, ECOG PS < 1, and the absence of brain metastases.
Collapse
Affiliation(s)
- Nargiz Majidova
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Mustafa Seyyar
- Department of Internal Medicine, Division of Medical Oncology, Kocaeli University, Kocaeli, Turkey
| | - Demet Işık Bayraktar
- Department of Internal Medicine, Division of Medical Oncology, Ondokuz Mayıs University Faculty of Medicine, Samsun, Turkey
| | - Gülhan Dinç
- Department of Internal Medicine, Division of Medical Oncology, Professor Dr Cemil Tascioglu City Hospital, Istanbul, Turkey
| | - Elfag İsgandarov
- Department of Internal Medicine, Division of Medical Oncology, Ondokuz Mayıs University Faculty of Medicine, Samsun, Turkey
| | - Javid Huseynov
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Alper Yaşar
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Abdussamet Çelebi
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Nadiye Sever
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Erkam Kocaaslan
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Pınar Erel
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Yeşim Ağyol
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Ali Kaan Güren
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Rukiye Arıkan
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Selver Işık
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Özlem Ercelep
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Güzin Demirağ
- Department of Internal Medicine, Division of Medical Oncology, Ondokuz Mayıs University Faculty of Medicine, Samsun, Turkey
| | - Umut Kefeli
- Department of Internal Medicine, Division of Medical Oncology, Kocaeli University, Kocaeli, Turkey
| | - Osman Köstek
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - İbrahim Vedat Bayoğlu
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| | - Murat Sarı
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Turkey
| |
Collapse
|
254
|
Heninger E, Breneman MT, Recchia EE, Kerr SC, Dogru RE, Sharifi MN, LeBeau AM, Kosoff D. Dynamic reciprocal interactions between activated T cells and tumor associated macrophages drive macrophage reprogramming and proinflammatory T cell migration within prostate tumor models. Sci Rep 2024; 14:24230. [PMID: 39414902 PMCID: PMC11484957 DOI: 10.1038/s41598-024-75265-9] [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: 07/16/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Tumor-associated macrophages (TAMs) have been implicated as a tumor microenvironment (TME) cell population, which may be playing a vital role in the inhibition of effective T cell responses in the prostate TME. In this manuscript, we leverage a novel microscale cell culture platform, known as Stacks, to investigate mono-, co-, and tri-culture TME models comprised of prostate tumor cell lines, primary macrophages, and autologous T cells from patients with prostate cancer. Through multiplexed analysis of these multi-cellular prostate tumor models, we capture a dynamic interaction between primary TAMs and activated T cells that resulted in reciprocal proinflammatory activation of both cell populations upon interaction. These findings suggest that activated T cells are capable of reprogramming immunosuppressive TAMs in the context of prostate tumor models and that TAM reprogramming may play a key supportive role in restoring proinflammatory T cell tumor responses in the prostate TME.
Collapse
Affiliation(s)
- Erika Heninger
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Sheena Catherine Kerr
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Reyna Elvan Dogru
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Aaron Matthew LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - David Kosoff
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S Middleton Memorial Veteran's Hospital, Madison, WI, USA.
- Department of Medicine, Carbone Cancer Center, University of Wisconsin Madison, 1111 Highland Avenue, WIMR 7105, Madison, WI, USA.
| |
Collapse
|
255
|
Santagata S, Trotta AM, D’Alterio C, Napolitano M, Rea G, Di Napoli M, Portella L, Ieranò C, Guardascione G, Coppola E, Caux C, Dubois B, Boyle HJ, Carles J, Rossetti S, Azzaro R, Feroce F, Perdonà S, Fordellone M, Bello AM, Califano D, Chiodini P, Pignata S, Scala S. KIR2DL2/DL3+NKs and Helios+Tregs in Peripheral Blood Predict Nivolumab Response in Patients with Metastatic Renal Cell Cancer. Clin Cancer Res 2024; 30:4755-4767. [PMID: 39167621 PMCID: PMC11474171 DOI: 10.1158/1078-0432.ccr-24-0729] [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: 03/08/2024] [Revised: 05/31/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
PURPOSE To identify predictive factors of nivolumab sensitivity, peripheral blood NKs and regulatory T-cell (Treg) were evaluated in patients with metastatic renal cell carcinoma (mRCC) enrolled in the REVOLUTION trial. EXPERIMENTAL DESIGN Fifty-seven mRCCs being treated with nivolumab, as at least second-line of therapy, and 62 healthy donors were longitudinally evaluated (0-1-3-6-12 months) for peripheral NKs and Tregs, phenotype, and function. Multivariable logistic regression was conducted to identify the independent predictors. The 0.632+ internal cross-validation was used to avoid overfitting. The best cutoff value based on a 3-month clinical response was applied to progression-free survival (PFS) and overall survival (OS). Kaplan-Meier curves for PFS and OS were produced. RESULTS At pretreatment, mRCCs displayed high frequency of NKp46+NKs, NKp30+NKs, KIR2DL1+NKs, KIR2DL2/DL3+NKs, and PD1+NKs with reduced NK degranulation as well as high frequency of Tregs, PD1+Tregs, Helios+Tregs, and ENTPD1+Tregs. Responder patients, identified as a clinical response after 3 months of treatment, presented at pretreatment significantly low CD3+, high KIR2DL2/DL3+NKs, high PD1+Tregs, and high Helios+Tregs. Upon multivariate analysis, only KIR2DL2/DL3NKs and Helios+Tregs held as independent predictors of nivolumab responsiveness. The KIR2DL2/DL3+NKs >35.3% identified patients with longer OS, whereas the Helios+Tregs >34.3% displayed significantly longer PFS. After 1-month of nivolumab, responder patients showed low CD3+, high NKs, KIR2DL2/DL3+NKs, and ICOS+Tregs. Among these subpopulations, CD3+ and KIR2DL2/DL3+NKs held as independent predictors of nivolumab efficacy. Low CD3+ (≤71%) was significantly associated with longer PFS, whereas high KIR2DL2/DL3+NKs (>23.3%) were associated with both PFS and OS. CONCLUSIONS Pretreatment evaluation of Helios+Tregs/KIR2DL2/DL3+NKs and 1-month posttreatment CD3+/ KIR2DL2/DL3+NKs will predict nivolumab response in mRCCs.
Collapse
Affiliation(s)
- Sara Santagata
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Anna Maria Trotta
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Crescenzo D’Alterio
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Maria Napolitano
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Giuseppina Rea
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Marilena Di Napoli
- Uro-Gynecological Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Luigi Portella
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Caterina Ieranò
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Giuseppe Guardascione
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Elisabetta Coppola
- Uro-Gynecological Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Christophe Caux
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France.
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France.
| | - Bertrand Dubois
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France.
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France.
| | - Helen J. Boyle
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France.
| | - Joan Carles
- Oncology Department, Val d’Hebron University, Barcelona, Spain.
| | - Sabrina Rossetti
- Uro-Gynecological Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Rosa Azzaro
- Transfusion Medicine Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Florinda Feroce
- Department of Pathology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Sisto Perdonà
- Department of Urology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Mario Fordellone
- Unità di Statistica Medica Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy.
| | - Anna Maria Bello
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Daniela Califano
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Paolo Chiodini
- Unità di Statistica Medica Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy.
| | - Sandro Pignata
- Uro-Gynecological Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
256
|
Lynch C, Arshad M, Katipally RR, Pitroda S, Weichselbaum R. Sharing the Burden: The Case for Definitive Local Therapy in Place of Immune Checkpoint Blockade for Patients With a Low-Volume Burden of Metastatic Disease. J Clin Oncol 2024; 42:3387-3391. [PMID: 39038267 PMCID: PMC11458364 DOI: 10.1200/jco.24.00549] [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: 03/14/2024] [Revised: 04/19/2024] [Accepted: 05/08/2024] [Indexed: 07/24/2024] Open
Abstract
COMMENTARY Sharing the burden of low-volume metastatic cancer between ICB and local treatments.
Collapse
Affiliation(s)
- Connor Lynch
- University of Chicago Medical Center, Department of Radiation and Cellular Oncology, 5758 S Maryland Ave, MC 9006, Chicago, IL 60637
| | - Muzamil Arshad
- University of Chicago Medical Center, Department of Radiation and Cellular Oncology, 5758 S Maryland Ave, MC 9006, Chicago, IL 60637
| | - Rohan R Katipally
- University of Chicago Medical Center, Department of Radiation and Cellular Oncology, 5758 S Maryland Ave, MC 9006, Chicago, IL 60637
| | - Sean Pitroda
- University of Chicago Medical Center, Department of Radiation and Cellular Oncology, 5758 S Maryland Ave, MC 9006, Chicago, IL 60637
| | - Ralph Weichselbaum
- University of Chicago Medical Center, Department of Radiation and Cellular Oncology, 5758 S Maryland Ave, MC 9006, Chicago, IL 60637
| |
Collapse
|
257
|
Huang J, Xiong L, Tang S, Zhao J, Zuo L. Balancing Tumor Immunotherapy and Immune-Related Adverse Events: Unveiling the Key Regulators. Int J Mol Sci 2024; 25:10919. [PMID: 39456702 PMCID: PMC11507008 DOI: 10.3390/ijms252010919] [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: 08/28/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Tumor immunotherapy has emerged as a promising approach in cancer treatment in recent years, offering vast potential. This method primarily involves targeting and inhibiting the suppressive checkpoints present in different immune cells to enhance their activation, ultimately leading to tumor regression. However, tumor cells exploit the surrounding immune cells and tissues to establish a tumor microenvironment (TME) that supports their survival and growth. Within the TME, the efficacy of effector immune cells is compromised, as tumor cells exploit inhibitory immune cells to suppress their function. Furthermore, certain immune cells can be co-opted by tumor cells to facilitate tumor growth. While significantly enhancing the body's tumor immunity can lead to tumor regression, it can also result in severe toxic side effects and an inflammatory factor storm. As a consequence, patients often discontinue treatment due to immune-related adverse events (irAEs) or, in extreme cases, succumb to toxic side effects before experiencing tumor regression. In this analysis, we examined several remission regimens for irAEs, each with its own drawbacks, including toxic side effects or suppression of tumor immunotherapy, which is undesirable. A recent research study, specifically aimed at downregulating intestinal epithelial barrier permeability, has shown promising results in reducing the severity of inflammatory bowel disease (IBD) while preserving immune function. This approach effectively reduces the severity of IBD without compromising the levels of TNF-α and IFN-γ, which are crucial for maintaining the efficacy of tumor immunotherapy. Based on the substantial similarities between IBD and ICI colitis (combo immune checkpoint inhibitors-induced colitis), this review proposes that targeting epithelial cells represents a crucial research direction for mitigating irAEs in the future.
Collapse
Affiliation(s)
- Jianshang Huang
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Lei Xiong
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Sainan Tang
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Junhao Zhao
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| | - Li Zuo
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Sciences, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China; (J.H.)
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No.81, Meishan Rd., Hefei 230032, China
| |
Collapse
|
258
|
Chang JY, Xu X, Shroff GS, Comeaux NI, Li W, Rodon Ahnert J, Karp DD, Dumbrava EE, Verma V, Chen A, Welsh J, Hong DS. Phase I/II study of BMS-986156 with ipilimumab or nivolumab with or without stereotactic ablative radiotherapy in patients with advanced solid malignancies. J Immunother Cancer 2024; 12:e009975. [PMID: 39384194 PMCID: PMC11474930 DOI: 10.1136/jitc-2024-009975] [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: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND BMS-986156 is an agonist of the glucocorticoid-induced tumor necrosis factor receptor (TNFR)-related protein (GITR) and promotes increased effector T-cell activation. Combined anti-GITR, anti-programmed death-1, anti-cytotoxic T-lymphocyte-associated protein 4 antibodies and radiotherapy improve tumor control in preclinical studies. Herein we describe the results of the safety and efficacy of BMS-986156+ipilimumab or nivolumab with/without stereotactic ablative radiotherapy (SABR) in patients with advanced solid cancers (NCT04021043). METHODS This open-label, multigroup, single-center phase I/II study enrolled patients with histologically-confirmed stage IV solid cancers resistant to standard treatments. Group 1 (G1, n=20) received four cycles of ipilimumab (3 mg/kg) plus BMS-986156 (30 mg as dose level 1 (L1) or 100 mg as dose level 2 (L2)), every 3 weeks (Q3W). Group 2 (G2, n=10) received four cycles of ipilimumab (3 mg/kg) plus BMS-986156 (dose as determined in G1, Q3W) with SABR (50 Gy/4 fx or 60-70 Gy/10 fx to liver/lung lesions. Group 3 (G3, n=20) received four cycles of nivolumab (480 mg) plus BMS-986156 (30 mg), every 4 weeks with SABR. Maintenance nivolumab could be given up to 2 years. Tumor responses were assessed every 1-3 months until progression, using immune-related response criteria. RESULTS 50 patients were enrolled between 10/2019 and 12/2021. Patients received a median of 3 (IQR 2-4.25) initial treatment cycles. 100 mg BMS-986156 with ipilimumab was tolerated well. Five discontinued BMS-986156 with ipilimumab due to treatment-related adverse events (TRAEs), with three in G1/L1, one in G1/L2 and one in G2, respectively. 22 patients (44%) experienced Grade 1-3 TRAEs (6, 4, 5, 7 patients for G1/L1, G1/L2, G2, G3). Six (12%) had Grade 3 TRAEs (2, 2, 1, 1 for G1/L1, G1/L2, G2, G3), with elevated alanine aminotransferase (n=3, in G1/L2, G2 and G3) and aspartate aminotransferase (n=2, in G2 and G3) being the most common. There was no Grade 4-5 TRAEs. Overall, 19/39 (48.7%) patients eligible for efficacy analysis had stable disease and 3 (7.7%) achieved a partial response. Out-of-field (abscopal) disease control rate (ACR) and out-of-field (abscopal) response rate (ARR) were 38.5% and 7.7%, respectively, with the highest ACR (50%, 9/18) and ARR (11.1%, 2/18) in G3. CONCLUSIONS BMS-986156 was well-tolerated with ipilimumab, nivolumab, with or without SABR. Outcomes were encouraging in this population, as more than half of patients had stable disease/partial response.
Collapse
Affiliation(s)
- Joe Y Chang
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xinyan Xu
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Girish S Shroff
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nathan I Comeaux
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Li
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jordi Rodon Ahnert
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ecaterina E Dumbrava
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vivek Verma
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Aileen Chen
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - James Welsh
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
259
|
Waddell T, Pillai M, Armitage K, Graham DM, Moran M, Dilleen M, Holmes S, Śleszyńska-Dopiera E, Hawkins R. Real-world effectiveness of first- and second-line anti-angiogenesis therapy in RCC: analysis of a UK-based population. Future Oncol 2024; 20:2547-2558. [PMID: 39382446 PMCID: PMC11534105 DOI: 10.1080/14796694.2024.2385882] [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: 08/23/2023] [Accepted: 07/27/2024] [Indexed: 10/10/2024] Open
Abstract
Aim: Renal cell carcinoma (RCC) is the seventh commonest cancer in the UK, where first-line (1L) sunitinib and second-line (2L) axitinib are treatment options.Methods: Retrospective, non-interventional data from the Christie NHS Foundation Trust (Manchester, UK). The primary end point was median progression-free survival (mPFS).Results: For 1L sunitinib (n = 622) and 2L axitinib (n = 121), mPFS (95% CI) was 8.4 (7.6, 9.9) and 6.2 (4.9, 9.3) months, respectively. In 1L, Karnofsky performance status, lactate dehydrogenase (LDH), neutrophils, hemoglobin, time from diagnosis to treatment and age were predictors (p < 0.05) of PFS. In 2L, LDH and platelets were predictors of PFS (p < 0.05).Conclusion: Sunitinib and axitinib were effective treatments for RCC. PFS predictors varied between 1L and 2L; LDH was a predictor for both.Clinical Trial Registration: NCT04033991 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Tom Waddell
- Christie NHS Founsdation Trust, Medical Oncology, Manchester, UK
| | - Manon Pillai
- Christie NHS Founsdation Trust, Medical Oncology, Manchester, UK
| | - Kate Armitage
- Christie NHS Founsdation Trust, Medical Oncology, Manchester, UK
| | - Donna M Graham
- Christie NHS Founsdation Trust, Medical Oncology, Manchester, UK
- University of Manchester, Manchester, UK
| | | | | | | | | | - Robert Hawkins
- Christie NHS Founsdation Trust, Medical Oncology, Manchester, UK
- University of Manchester, Manchester, UK
| |
Collapse
|
260
|
Stransky LA, Gao W, Schmidt LS, Bi K, Ricketts CJ, Ramesh V, James A, Difilippantonio S, Ileva L, Kalen JD, Karim B, Jeon A, Morgan T, Warner AC, Turan S, Unite J, Tran B, Choudhari S, Zhao Y, Linn DE, Yun C, Dhandapani S, Parab V, Pinheiro EM, Morris N, He L, Vigeant SM, Pignon JC, Sticco-Ivins M, Signoretti S, Van Allen EM, Linehan WM, Kaelin WG. Toward a CRISPR-based mouse model of Vhl-deficient clear cell kidney cancer: Initial experience and lessons learned. Proc Natl Acad Sci U S A 2024; 121:e2408549121. [PMID: 39365820 PMCID: PMC11474080 DOI: 10.1073/pnas.2408549121] [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: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024] Open
Abstract
CRISPR is revolutionizing the ability to do somatic gene editing in mice for the purpose of creating new cancer models. Inactivation of the VHL tumor suppressor gene is the signature initiating event in the most common form of kidney cancer, clear cell renal cell carcinoma (ccRCC). Such tumors are usually driven by the excessive HIF2 activity that arises when the VHL gene product, pVHL, is defective. Given the pressing need for a robust immunocompetent mouse model of human ccRCC, we directly injected adenovirus-associated viruses (AAVs) encoding sgRNAs against VHL and other known/suspected ccRCC tumor suppressor genes into the kidneys of C57BL/6 mice under conditions where Cas9 was under the control of one of two different kidney-specific promoters (Cdh16 or Pax8) to induce kidney tumors. An AAV targeting Vhl, Pbrm1, Keap1, and Tsc1 reproducibly caused macroscopic ccRCCs that partially resembled human ccRCC tumors with respect to transcriptome and cell of origin and responded to a ccRCC standard-of-care agent, axitinib. Unfortunately, these tumors, like those produced by earlier genetically engineered mouse ccRCCs, are HIF2 independent.
Collapse
Affiliation(s)
- Laura A. Stransky
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Wenhua Gao
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Laura S. Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Kevin Bi
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02115
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA02115
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - Christopher J. Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
| | - Vijyendra Ramesh
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Amy James
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Simone Difilippantonio
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Lilia Ileva
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Joseph D. Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Albert Jeon
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Tamara Morgan
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Andrew C. Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Sevilay Turan
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Joanne Unite
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Bao Tran
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Sulbha Choudhari
- Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD21701
| | - Yongmei Zhao
- Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD21701
| | | | - Changhong Yun
- Pharmacokinetics, Merck & Co., Inc., Boston, MA02115
| | | | - Vaishali Parab
- Pharmacokinetics, Merck & Co., Inc., South San Francisco, CA94080
| | | | - Nicole Morris
- Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Lixia He
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Sean M. Vigeant
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Jean-Christophe Pignon
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
| | - Maura Sticco-Ivins
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA02115
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA02115
| | - Eliezer M. Van Allen
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02115
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA02115
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - W. Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
| | - William G. Kaelin
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
261
|
Rodriguez SMB, Tataranu LG, Kamel A, Turliuc S, Rizea RE, Dricu A. Glioblastoma and Immune Checkpoint Inhibitors: A Glance at Available Treatment Options and Future Directions. Int J Mol Sci 2024; 25:10765. [PMID: 39409094 PMCID: PMC11477435 DOI: 10.3390/ijms251910765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma is known to be one of the most aggressive and fatal human cancers, with a poor prognosis and resistance to standard treatments. In the last few years, many solid tumor treatments have been revolutionized with the help of immunotherapy. However, this type of treatment has failed to improve the results in glioblastoma patients. Effective immunotherapeutic strategies may be developed after understanding how glioblastoma achieves tumor-mediated immune suppression in both local and systemic landscapes. Biomarkers may help identify patients most likely to benefit from this type of treatment. In this review, we discuss the use of immunotherapy in glioblastoma, with an emphasis on immune checkpoint inhibitors and the factors that influence clinical response. A Pubmed data search was performed for all existing information regarding immune checkpoint inhibitors used for the treatment of glioblastoma. All data evaluating the ongoing clinical trials involving the use of ICIs either as monotherapy or in combination with other drugs was compiled and analyzed.
Collapse
Affiliation(s)
- Silvia Mara Baez Rodriguez
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Amira Kamel
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy “G. T. Popa”, 700115 Iasi, Romania;
| | - Radu Eugen Rizea
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania;
| |
Collapse
|
262
|
Ide T, Araki T, Koizumi T. Thromboembolism during immune checkpoint inhibitor therapy: frequency and risk factors. Discov Oncol 2024; 15:527. [PMID: 39367999 PMCID: PMC11455762 DOI: 10.1007/s12672-024-01416-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/01/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Thromboembolism (TE) is a well-known complication during chemotherapy in cancer patients. However, the risk of TE associated with immune checkpoint inhibitors (ICIs) is unknown. This study was performed to investigate the incidence of TE and associated risk factors in patients treated with ICIs. METHODS We conducted a retrospective chart survey of patients receiving at least one ICI at Shinshu University Hospital between September 2014 and October 2021. Age, sex, cancer type, body mass index, medical history, laboratory data at commencement of treatment, and medication data were obtained from electronic medical records. TE events (venous thromboembolism [VTE], arterial thromboembolism [ATE]) were identified after ICI initiation. RESULTS The study population consisted of 548 patients with a median age of 70.0 (19-89) years, 71.4% men, and a median follow-up of 15.1 months (range; 0.16-72.0 months). Nivolumab was the most commonly used ICI (45.8%), followed by pembrolizumab (23.9%), pembrolizumab plus anticancer drugs (7.8%), and nivolumab plus ipilimumab (5.1%). Thirty-eight cases of TE (6.9%) occurred (22 VTE, 16 ATE). Risk factors significantly associated with TE in multivariate logistic analysis were dyslipidemia (OR 2.44; 95% CI 1.17-5.09; p = 0.017), Khorana score ≥ 2 (HR 2.40; 95% CI 1.14-5.04; p = 0.021). Overall survival was not significantly different from patients without TE (p = 0.963). CONCLUSION These results suggested that the frequency of TE is higher than expected and should be considered and monitored in patients treated with ICIs.
Collapse
Affiliation(s)
- Takayuki Ide
- Department of Pharmacy, Shinshu University Hospital, 3-1-1 Asahi Matsumoto-Shi, Nagano, 390-8621, Japan
| | - Taisuke Araki
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi Matsumoto-Shi, Nagano, 390-8621, Japan
| | - Tomonobu Koizumi
- Department of Hematology and Medical Oncology, Shinshu University School of Medicine, 3-1-1 Asahi Matsumoto-Shi, Nagano, 390-8621, Japan.
| |
Collapse
|
263
|
Panian J, Saidian A, Hakimi K, Ajmera A, Anderson WJ, Barata P, Berg S, Signoretti S, Lee Chang S, D’Andrea V, George D, Dzimitrowicz H, El Zarif T, Emamekhoo H, Gross E, Kilari D, Lam E, Lashgari I, Psutka S, Rauterkus GP, Shabaik A, Thapa B, Wang L, Weise N, Yim K, Zhang T, Derweesh I, McKay RR. Pathological Outcomes of Patients With Advanced Renal Cell Carcinoma Who Receive Nephrectomy Following Immunotherapy. Oncologist 2024; 29:870-877. [PMID: 37368355 PMCID: PMC11448883 DOI: 10.1093/oncolo/oyad166] [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: 12/10/2022] [Accepted: 01/31/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Even though cytoreductive nephrectomy (CN) was once the standard of care for patients with advanced renal cell carcinoma (RCC), its role in treatment has not been well analyzed or defined in the era of immunotherapy (IO). MATERIALS AND METHODS This study analyzed pathological outcomes in patients with advanced or metastatic RCC who received IO prior to CN. This was a multi-institutional, retrospective study of patients with advanced or metastatic RCC. Patients were required to receive IO monotherapy or combination therapy prior to radical or partial CN. The primary endpoint assessed surgical pathologic outcomes, including American Joint Committee on Cancer (AJCC) staging and frequency of downstaging, at the time of surgery. Pathologic outcomes were correlated to clinical variables using a Wald-chi squared test from Cox regression in a multi-variable analysis. Secondary outcomes included objective response rate (ORR) defined by response evaluation criteria in solid tumors (RECIST) version 1.1 and progression-free survival (PFS), which were estimated using the Kaplan-Meier method with reported 95% CIs. RESULTS Fifty-two patients from 9 sites were included. Most patients were male (65%), 81% had clear cell histology, 11% had sarcomatoid differentiation. Overall, 44% of patients experienced pathologic downstaging, and 13% had a complete pathologic response. The ORR immediately prior to nephrectomy was stable disease in 29% of patients, partial response in 63%, progressive disease in 4%, and 4% unknown. Median follow-up for the entire cohort was 25.3 months and median PFS was 3.5 years (95% CI, 2.1-4.9). CONCLUSIONS IO-based interventions prior to CN in patients with advanced or metastatic RCC demonstrates efficacy, with a small fraction of patients showing a complete response. Additional prospective studies are warranted to investigate the role of CN in the modern IO-era.
Collapse
Affiliation(s)
- Justine Panian
- University of California San Diego, Department of Medicine, Division of Hematology-Oncology La Jolla, CA, USA
| | - Ava Saidian
- University of California San Diego, Department of Urology, La Jolla, CA, USA
| | - Kevin Hakimi
- University of California San Diego, Department of Urology, La Jolla, CA, USA
| | - Archana Ajmera
- University of California San Diego, Department of Medicine, Division of Hematology-Oncology, La Jolla, CA, USA
| | | | - Pedro Barata
- Tulane University, Deming Department of Medicine, New Orleans, LA, USA
| | - Stephanie Berg
- Loyola University Chicago, Department of Cancer Biology and Internal Medicine, Maywood, IL, USA
| | - Sabina Signoretti
- Brigham and Women’s Hospital, Department of Pathology, Boston, MA, USA
| | - Steven Lee Chang
- Brigham and Women’s Hospital, Division of Urology, Boston, MA, USA
| | - Vincent D’Andrea
- Brigham and Women’s Hospital, Division of Urology, Boston, MA, USA
| | - Daniel George
- Duke Cancer Institute, Department of Medicine, Durham, NC, USA
| | | | - Talal El Zarif
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA
| | - Hamid Emamekhoo
- University of Wisconsin, Department of Medicine, Madison, WI, USA
| | - Evan Gross
- The University of Washington, Department of Urology, Seattle, WA, USA
| | - Deepak Kilari
- Medical College of Wisconsin, Department of Internal Medicine, Milwaukee, WI, USA
| | - Elaine Lam
- University of Colorado Cancer Center, Division of Medical Oncology, Aurora, CO, USA
| | - Isabel Lashgari
- San Diego State University, Department of Cell and Molecular Biology, San Diego, CA, USA
| | - Sarah Psutka
- The University of Washington, Department of Urology, Seattle, WA, USA
| | - Grant P Rauterkus
- Tulane University, Deming Department of Medicine, New Orleans, LA, USA
| | - Ahmed Shabaik
- University of California San Diego, Department of Pathology, La Jolla, CA, USA
| | - Bicky Thapa
- Medical College of Wisconsin, Department of Internal Medicine, Milwaukee, WI, USA
| | - Luke Wang
- University of California San Diego, Department of Urology, La Jolla, CA, USA
| | - Nicole Weise
- University of California San Diego, Department of Medicine, Division of Hematology-Oncology La Jolla, CA, USA
| | - Kendrick Yim
- Brigham and Women’s Hospital, Division of Urology, Boston, MA, USA
| | - Tian Zhang
- UT Southwestern, Department of Internal Medicine, Dallas, TX, USA
| | - Ithaar Derweesh
- University of California San Diego, Department of Urology, La Jolla, CA, USA
| | - Rana R McKay
- University of California San Diego, Department of Medicine, Department of Urology, La Jolla, CA, USA
| |
Collapse
|
264
|
Osawa T, Sasaki K, Machida R, Matsumoto T, Matsui Y, Kitamura H, Nishiyama H. Real-world treatment trends for patients with advanced prostate cancer and renal cell carcinoma and their cost-a survey in Japan. Jpn J Clin Oncol 2024; 54:1062-1070. [PMID: 38843876 DOI: 10.1093/jjco/hyae045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/28/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Advanced (Stage IV) prostate and renal cancer have poor prognosis, and several therapies have been developed, but many are very costly. This study investigated drug regimens used in patients with untreated Stage IV prostate cancer and renal cell carcinoma and calculated the monthly cost of each. METHODS We surveyed first-line drugs administered to patients with untreated Stage IV prostate cancer and renal cancer at Japan Clinical Oncology Group affiliated centers from April 2022 to March 2023. Drug costs were calculated according to drug prices in September 2023. Individual drug costs were calculated or converted to 28-day costs. RESULTS A total of 700 patients with untreated Stage IV prostate cancer were surveyed. Androgen deprivation therapy + androgen receptor signaling inhibitor was the most common regimen (56%). The cost of androgen deprivation therapy + androgen receptor signaling inhibitor was 10.6-30.8-fold compared with conventional treatments. A total of 137 patients with Stage IV renal cancer were surveyed. Among them, 91% of patients received immune-oncology drug-based regimen. All patients received treatments with a monthly cost of ≥500 000 Japanese yen, and 80.4% of patients received treatments with a monthly cost of ≥1 million Japanese yen, of combination treatments. The cost of immune-oncology drug-based regimen was 1.2-3.1-fold that of TKI alone. CONCLUSION To the best of our knowledge, this is the first report of a survey of first-line drug therapy in untreated Stage IV prostate cancer and renal cell carcinoma stratified by age and treatment costs. Our results show that most Japanese patients received state-of-the-art, effective treatments with high financial burden.
Collapse
Affiliation(s)
- Takahiro Osawa
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Keita Sasaki
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Ryunosuke Machida
- Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Matsumoto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshiyuki Matsui
- Department of Urology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroshi Kitamura
- Department of Urology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Hiroyuki Nishiyama
- Department of Urology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
265
|
Nerli RB, Gautam S, Ghagane SC, Rai S. Impact of COVID-19 on Treatment in Patients with Renal Cell Carcinoma. ARCHIVES OF RAZI INSTITUTE 2024; 79:1091-1097. [PMID: 40292050 PMCID: PMC12018751 DOI: 10.32592/ari.2024.79.5.1091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/02/2023] [Indexed: 04/30/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The current pandemic has resulted in a significant reallocation of health-care resources, with the recommended treatment strategy advocating for oncology patients was to delay elective procedures. A retrospective analysis was conducted to evaluatethe impact of COVID-19 on patients with renal cell carcinoma (RCC) and the associated treatment protocols. A retrospective review of the inpatient and outpatient records of all patients presenting with renal cell carcinoma during the period from March 2020 to the end of March 2021 was conducted. A total of 26 patients (21 males and 5 females) with a mean age of 55.46±9.44 years were diagnosed with an operable renal mass during the study period. The mean hospitalisation period (15.19±2.28) was found to be longer in patients who required surgical intervention. The delay was attributable to a number of factors, including the necessity for pre-operative testing using RT-PCR, a chest HR-CT, clearance from the chest physician, and preparation. The overall cost of hospitalization increased in these patients compared to the pre-pandemic period due to a number of factors, including prolonged hospitalization, an increased incidence of complications, the necessity for pre-operative testing for SARS-CoV-2, the use of personal protective equipment, and the provision of nursing care. During the same period, three out of eight patients who had metastatic disease with positive RT-PCR were initiated on targeted therapy, while the remaining underwent cytoreductive nephrectomy. The study concludes that patients with RCC seeking treatment during the current pandemic face significant challenges, including delays in treatment, increased hospitalization rates, and a rise in testing, which collectively contribute to elevated treatment costs. It is imperative to conduct a long-term follow-up to ascertain whether these factors have influenced the outcome of the patients in question.
Collapse
Affiliation(s)
- R B Nerli
- Department of Urology, JN Medical College, KLE Academy of Higher Education & Research (Deemed-to-be-University), JNMC Campus, Nehru Nagar, Belagavi-590010, Karnataka, India
| | - S Gautam
- Department of Respiratory Medicine, JN Medical College, KLE Academy of Higher Education & Research (Deemed-to-be-University), JNMC Campus, Nehru Nagar, Belagavi-590010, Karnataka, India
| | - S C Ghagane
- KAHER's Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education & Research (Deemed-to-be-University), JNMC Campus, Nehru Nagar, Belagavi-590010, Karnataka, India
| | - S Rai
- Department of Urology, JN Medical College, KLE Academy of Higher Education & Research (Deemed-to-be-University), JNMC Campus, Nehru Nagar, Belagavi-590010, Karnataka, India
| |
Collapse
|
266
|
Zhang Y, Huang Y, Yu D, Xu M, Hu H, Zhang Q, Cai M, Geng X, Zhang H, Xia J, Guo M, Lu D, Xu H, Li L, Zhang X, Wang Q, Liu S, Zhang W. Demethylzeylasteral induces PD-L1 ubiquitin-proteasome degradation and promotes antitumor immunity via targeting USP22. Acta Pharm Sin B 2024; 14:4312-4328. [PMID: 39525573 PMCID: PMC11544192 DOI: 10.1016/j.apsb.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/11/2024] [Accepted: 07/25/2024] [Indexed: 11/16/2024] Open
Abstract
Programmed cell death ligand-1 (PD-L1) is a T cell inhibitory immune checkpoint molecule that interacts with programmed cell death-1 (PD-1) to promote immune escape of tumor cells. Compared with antibody therapies, small molecule drugs show better prospects due to their advantages such as higher bioavailability, better tissue penetration, and reduced risk of immunogenicity. Here, we found that the small molecule demethylzeylasteral (Dem) can significantly downregulate the expression of PD-L1 in colorectal cancer cells and enhance the killing effect of T cells on tumor cells. Mechanistically, Dem binds to the deubiquitinating enzyme USP22 and promotes its degradation, resulting in increased ubiquitination and degradation of PD-L1 through the proteasome pathway. In addition, Dem increased the activity of cytotoxic T cells and reduced the number of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) in tumor-infiltrating lymphocytes (TILs), thereby activating the tumor immune microenvironment and inhibiting the growth of subcutaneous MC38 tumors in C57BL/6 mice. Moreover, we also found that the combination of Dem and CTLA4 antibodies can further improve the efficacy of antitumor therapy. Our study reveals the mechanism by which Dem promotes PD-L1 degradation and suggests that the combination of Dem and CTLA4 antibodies may improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Yanyan Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yun Huang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dianping Yu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mengting Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongmei Hu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qing Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Minchen Cai
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiangxin Geng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongwei Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianhua Xia
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mengmeng Guo
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dong Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hanchi Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Linyang Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xing Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qun Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
267
|
Zhou C, Jiang J, Xiang X, Liu H, Wu G, Zeng R, Lu T, Zhang M, Shen Y, Hong M, Zhang J. Preclinical investigations and a first-in-human phase 1a trial of JS007, a novel anti-CTLA-4 antibody, in patients with advanced solid tumors. Exp Hematol Oncol 2024; 13:98. [PMID: 39354625 PMCID: PMC11443874 DOI: 10.1186/s40164-024-00567-7] [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: 06/24/2024] [Accepted: 09/21/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Blocking cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) shows substantial antitumor efficacy. Here, we report the preclinical data and outcomes of a first-in-human phase 1a trial of JS007, a novel anti-CTLA-4 antibody, in advanced solid tumors. METHODS In preclinical studies, both in vitro characteristics and in vivo characteristics of JS007 were investigated. The clinical trial included a dose escalation phase and a dose expansion phase. Eligible patients with previously treated advanced solid tumors were enrolled. In the dose escalation phase, JS007 was administered intravenously every 3 weeks at doses of 0.03, 0.3, 1, 3, and 10 mg/kg. Then, 3 and 10 mg/kg were chosen for the dose expansion phase. The primary endpoints included the maximum tolerated dose (MTD) of JS007 based on dose-limiting toxicities (DLTs) and safety. RESULTS JS007 could effectively bind to CTLA-4 and induce an immune response in vitro. Potent in vivo antitumor activity of JS007 was observed. Increased T cell infiltration and T regulatory (Treg) cell depletion in tumor microenvironment of cancer cell xenografts were detected after treated with JS007. Pharmacological analysis in experimental animals showed a dose-proportional increase in exposure. In the clinical trial, a total of 28 patients were treated with JS007 across 5 dose levels. No DLTs occurred. The MTD did not reach at the highest dose tested (10 mg/kg). Twenty-three (82.1%) patients experienced at least one treatment-related adverse event (TRAE). The incidence of Grade ≥ 3 TRAEs was 28.6% (8/28) with alanine aminotransferase increase (7.1%, 2/28) being the most frequently reported TRAE. No severe immune-related adverse event (irAE) occurred. Pharmacological profiles of JS007 in patients were similar to those in animal models. Serum concentration of JS007 showed a dose-dependent escalation, and the half-life of JS007 was 9.4 ~ 12.2 days. Treatment-induced anti-drug antibody was detected in 2 patients. The disease control rate was 50% (14/28), and the median overall survival was 14.7 months. CONCLUSIONS JS007 preliminarily demonstrates good tolerance and encouraging antitumor activity in patients with previously treated advanced solid tumors. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT05049265 (Sep 20, 2021).
Collapse
Affiliation(s)
- Chenfei Zhou
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinling Jiang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojun Xiang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guowu Wu
- Department of Medical Oncology, Cancer Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Ruichao Zeng
- Shanghai Junshi Biosciences Co., Ltd, Shanghai, China
| | - Tong Lu
- Shanghai Junshi Biosciences Co., Ltd, Shanghai, China
| | - Mengqi Zhang
- Shanghai Junshi Biosciences Co., Ltd, Shanghai, China
| | - Yuteng Shen
- Shanghai Junshi Biosciences Co., Ltd, Shanghai, China
| | - Min Hong
- Shanghai Junshi Biosciences Co., Ltd, Shanghai, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
268
|
Maksimovic S, Boscolo NC, La Posta L, Barrios S, Moussa MJ, Gentile E, Pesquera PI, Li W, Chen J, Gomez JA, Basi A, Burks JK, Alvarez-Breckenridge C, Gao J, Campbell MT, Dondossola E. Antiangiogenic Tyrosine Kinase Inhibitors have Differential Efficacy in Clear Cell Renal Cell Carcinoma in Bone. CANCER RESEARCH COMMUNICATIONS 2024; 4:2621-2637. [PMID: 39248577 PMCID: PMC11459607 DOI: 10.1158/2767-9764.crc-24-0304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent kidney neoplasm; bone metastasis (BM) develops in 35% to 40% of metastatic patients and results in substantial morbidity and mortality, as well as medical costs. A key feature of ccRCC is the loss of function of the von Hippel-Lindau protein, which enhances angiogenesis via vascular endothelial growth factor release. Consequently, antiangiogenic tyrosine kinase inhibitors (TKI) emerged as a treatment for ccRCC. However, limited data about their efficacy in BM is available, and no systematic comparisons have been performed. We developed mouse models of bone and lung ccRCC tumors and compared their anticancer efficacy, impact on mouse survival, and mechanisms of action, including effects on tumor cells and both immune and nonimmune (blood vessels and osteoclasts) bone stromal components. This approach elucidates the efficacy of TKIs in ccRCC bone tumors to support rational interrogation and development of therapies. SIGNIFICANCE TKIs showed different efficacy in synchronous bone and lung metastases and did not eradicate tumors as single agents but induced extensive reprogramming of the BM microenvironment. This resulted in a significant decrease in neoangiogenic blood vessels, bone remodeling, and immune cell infiltration (including CD8 T cells) with altered spatial distribution.
Collapse
Affiliation(s)
- Stefan Maksimovic
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Nina C. Boscolo
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ludovica La Posta
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Sergio Barrios
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Bioengineering, Rice University, Houston, Texas.
| | - Mohammad Jad Moussa
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Emanuela Gentile
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Pedro I. Pesquera
- Division of Surgery, Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Wenjiao Li
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jianfeng Chen
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Javier A. Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Akshay Basi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Jianjun Gao
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Matthew T. Campbell
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
269
|
Au KM, Wilson JE, Ting JPY, Wang AZ. An injectable subcutaneous colon-specific immune niche for the treatment of ulcerative colitis. Nat Biomed Eng 2024; 8:1243-1265. [PMID: 38049469 DOI: 10.1038/s41551-023-01136-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 10/14/2023] [Indexed: 12/06/2023]
Abstract
As a chronic autoinflammatory condition, ulcerative colitis is often managed via systemic immunosuppressants. Here we show, in three mouse models of established ulcerative colitis, that a subcutaneously injected colon-specific immunosuppressive niche consisting of colon epithelial cells, decellularized colon extracellular matrix and nanofibres functionalized with programmed death-ligand 1, CD86, a peptide mimic of transforming growth factor-beta 1, and the immunosuppressive small-molecule leflunomide, induced intestinal immunotolerance and reduced inflammation in the animals' lower gastrointestinal tract. The bioengineered colon-specific niche triggered autoreactive T cell anergy and polarized pro-inflammatory macrophages via multiple immunosuppressive pathways, and prevented the infiltration of immune cells into the colon's lamina propria, promoting the recovery of epithelial damage. The bioengineered niche also prevented colitis-associated colorectal cancer and eliminated immune-related colitis triggered by kinase inhibitors and immune checkpoint blockade.
Collapse
Affiliation(s)
- Kin Man Au
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Justin E Wilson
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew Z Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
270
|
Sato K, Sazuka T, Arai T, Sato H, Kanesaka M, Ando K, Saito S, Pae S, Yamada Y, Imamura Y, Sakamoto S, Ichikawa T. Machine learning analysis for detecting late recurrence and loss to follow-up after renal cell carcinoma surgery. BJUI COMPASS 2024; 5:950-956. [PMID: 39416750 PMCID: PMC11479800 DOI: 10.1002/bco2.425] [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: 05/01/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives Renal cell carcinoma (RCC) is shown to have a tendency for late recurrence, occurring 5 or more years after curative surgery. Imaging diagnosis is required for follow-up, and there is no definitive answer as to how long this should continue. Some patients discontinue follow-up visits at their own discretion. How best to predict late recurrence and loss to follow-up (LF) remains unclear. Patients and methods This study targeted patients diagnosed with non-metastatic RCC who underwent either radical or partial nephrectomy at Chiba University Hospital between 1988 and 2021. Follow-up for patients with RCC is typically lifelong. We used random survival forests (RSFs), a machine learning-based survival analysis method, to predict late recurrence and LF. For verification of prediction accuracy, we applied the time-dependent area under the receiver operating characteristic curve (t-AUC). To analyse the risks of late recurrence and LF, SurvSHAP(t) and partial dependence plots were used. Results We analysed 1051 cases in this study. Median follow-up was 58.5 (range: 0-376) months. The predictive accuracy of recurrence using RSF was t-AUC 0.806, 0.761, 0.674 and 0.566 at 60, 120, 180 and 240 months postoperatively, respectively. The recurrence risk impact showed a time-dependent increase up to approximately 50 months postoperatively. Beyond 50 months, there were no distinct risk factors characteristic of late recurrence. The predictive accuracy of LF using RSF was t-AUC 0.542, 0.699, 0.685, 0.628 and 0.674 at 60, 120, 180, 240 and 300 months postoperatively, respectively. The risk of LF increased with advancing age beyond 70 years. Conclusion It is difficult to identify factors that predict late recurrence. For long-term follow-up observation, it is essential to pay particular attention to patients with RCC aged 70 years and above. Establishing frameworks to facilitate collaboration with local hospitals near patients' residences and providing care within the community is necessary.
Collapse
Affiliation(s)
- Kodai Sato
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Tomokazu Sazuka
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Takayuki Arai
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Hiroaki Sato
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Manato Kanesaka
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Keisuke Ando
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Shinpei Saito
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Sangjon Pae
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Yasutaka Yamada
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Yusuke Imamura
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Shinichi Sakamoto
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Tomohiko Ichikawa
- Department of Urology, Graduate School of MedicineChiba UniversityChibaJapan
| |
Collapse
|
271
|
Zhu D, Huang Z, Xiao K, Bian D, Zhang J, Zhang Z, Zhu S, Yin L, Zhu J, Zhang P. Does the number of cycles of neoadjuvant therapy affect the efficacy of neoadjuvant chemoimmunotherapy for non-small cell lung cancer in locally advanced stage? Retrospective experience based on a single center. Asia Pac J Clin Oncol 2024; 20:643-651. [PMID: 37313687 DOI: 10.1111/ajco.13971] [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: 01/13/2023] [Revised: 04/30/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND The number of cycles of neoadjuvant therapy programmed cell death 1 (PD-1) inhibitor for locally advanced non-small cell lung cancer (NSCLC) remains controversial. METHODS From October 2019 to March 2022, neoadjuvant chemoimmunotherapy followed by radical surgery for NSCLC patients with stage II-III were retrospectively reviewed in Shanghai Pulmonary Hospital. The radiologic response was assessed according to the Response Evaluation Criteria for Solid Tumors version 1.1. The major pathological response was defined as no more than 10% residual tumor. Student's t-test, chi-square test, and Mann-Whitney test were used for univariate analysis, logistic regression analysis was used for multivariate analysis. All statistical analyses were calculated by SPSS software (version 26). RESULTS Among 108 patients, the number of patients who received 2-cycle (2-cycle group) and more than 2-cycle (>2-cycle group) neoadjuvant chemoimmunotherapy were 75 (69.4%) and 33 (30.6%), respectively. Compared with patients in the >2-cycle group, patients in the 2-cycle group had significantly smaller diagnostic radiological tumor size (37.0 mm vs. 49.6 mm, p = 0.022) and radiological tumor regression rate (36% vs. 49%, p = 0.007). However, no significant difference in pathological tumor regression rate was observed between patients in the 2-cycle group and >2-cycle group. Further logistic regression analysis demonstrated that the neoadjuvant chemoimmunotherapy cycle could independently affect the radiographic response (odds ratio [OR]: 0.173, 95% confidence interval [CI]: 0.051-0.584, p = 0.005) but not for pathological response (OR: 0.450, 95% CI: 0.161-1.257, p = 0.127). CONCLUSIONS For patients diagnosed with stage II-III NSCLC, the number of neoadjuvant cycles administered can significantly influence the radiographic efficacy of chemoimmunotherapy.
Collapse
Affiliation(s)
- Di Zhu
- School of Medicine, Shihezi University, Shihezi, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhida Huang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kai Xiao
- School of Medicine, Shihezi University, Shihezi, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongliang Bian
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhonghong Zhang
- Department of Respiration Department II, First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Shuncang Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Laibo Yin
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Jialong Zhu
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Peng Zhang
- School of Medicine, Shihezi University, Shihezi, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
272
|
Seront E, Reichel C, Lhommel R, Tombal B. A Case Series Study of the Role of Prostate-specific Membrane Antigen Positron Emission Tomography/Computed Tomography in Early Evaluation of the Response to Systemic Therapy in Metastatic Renal Cancer. Eur Urol Oncol 2024; 7:1147-1149. [PMID: 38453597 DOI: 10.1016/j.euo.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/01/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Affiliation(s)
- Emmanuel Seront
- Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Medical Oncology, Centre Universitaire HELORA, Haine Saint Paul, Belgium.
| | - Cédric Reichel
- Department of Nuclear Medicine, Centre Universitaire HELORA, Haine Saint Paul, Belgium
| | - Renaud Lhommel
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bertrand Tombal
- Department of Urology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
273
|
Silberg M, Krabbe LM, Bögemann M, Schrader AJ, Tully K, Schlack K. Immune-Related Adverse Events Can Predict Progression-Free and Overall Survival In Patients With Metastatic Renal Cell Carcinoma Treated With Immune Checkpoint Inhibitors. Clin Genitourin Cancer 2024; 22:102164. [PMID: 39153900 DOI: 10.1016/j.clgc.2024.102164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Different combination therapies using anti - PD-1 / PD-L1 or CTLA-4 immune checkpoint inhibition (ICI) are widely used in patients with metastatic renal cell carcinoma (mRCC). In the absents of established biomarkers, immune-related adverse events (irAEs) have been discussed as potential predictors of response. METHODS In this retrospective cohort study, data of 134 patients with mRCC undergoing ICI treatment (Nivolumab, Ipilimumab and Nivolumab, Pembrolizumab and Axitinib or Avelumab and Axitinib) between 2015 and 2021 were analyzed. To examine the utility of irAEs as predictors of overall survival (OS) and progression-free survival (PFS), separate Kaplan-Meier analyses and Cox proportional regression analyses were applied. Landmark analysis was conducted after 12 weeks to reduce immortal time bias. RESULT irAEs were observed in 85 patients (63.4%). Cutaneous (n = 52, 38.8%), endocrine (n = 33, 24.6%) and hepatic (n = 19, 14.2%) irAEs were most commonly observed. In Kaplan-Meier analysis, patients experiencing irAEs showed favorable median PFS (15 months, 95% CI, 9.91-20.09) compared to the non-irAE group (5 months, 95% CI, 3.56-6.44, P < .001). The median OS was 25 months (95% CI, 16.79-33.21) in the non-irAE group, while it was not reached in the irAE group (P = .002). In multivariable analysis, the presence of any irAE was associated with favorable PFS (HR 0.46 [95% CI, 0.26-0.82] P = .008) and OS (HR: 0.28 [95% CI, 0.12-0.63] P = .002), respectively. Landmark analysis after 12 weeks showed mixed results depending on the classification of the irAE group at the landmark time. CONCLUSION The presence of irAEs under ICI therapy in patients with mRCC is associated with better PFS and OS. Thus, manageable irAEs should not be cause for premature discontinuation of ICI therapy, as they seem to indicate favorable outcomes. Considering the time-dependent nature of irAEs is crucial estimating their value as predictive markers.
Collapse
Affiliation(s)
- Matteo Silberg
- Department of Urology, Marien Hospital Herne, University Hospital of the Ruhr University of Bochum, Herne, Germany.
| | | | - Martin Bögemann
- Department of Urology, University Hospital Muenster, Muenster, Germany
| | | | - Karl Tully
- Department of Urology, Marien Hospital Herne, University Hospital of the Ruhr University of Bochum, Herne, Germany
| | - Katrin Schlack
- Department of Urology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
274
|
Kührer N, Haas G, Wafa H, Klinglmair G, Pichler R. Sudden Vision Loss in a Patient With Renal Cell Carcinoma: A Potential Indicator of Choroidal Metastasis. Cureus 2024; 16:e71138. [PMID: 39391258 PMCID: PMC11465728 DOI: 10.7759/cureus.71138] [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] [Accepted: 10/09/2024] [Indexed: 10/12/2024] Open
Abstract
Choroidal metastasis of renal cell cancer (RCC) is an exceptionally rare clinical occurrence. In most cases, sudden vision loss is the first symptom. Here we present the case of a 52-year-old male with papillary RCC first diagnosed in 2018. Three years later, a bronchoscopy with endobronchial ultrasound (EBUS) identified suspicious infracarinal lymph nodes. A biopsy confirmed metastasis from RCC, leading to the initiation of systemic first-line therapy with cabozantinib monotherapy. The patient confirmed an excellent response with complete remission. In 2023, the patient reported for the first time a bilateral decrease in vision. Initial management with corrective lenses was unsuccessful. Further staging indicated metastases in the liver, spleen, and adrenal gland. Consecutively, the therapy was switched to lenvatinib and pembrolizumab. Two months later, an ophthalmologic examination due to persisting vision loss confirmed bilateral choroidal metastases. The systemic therapy was continued, and the patient's vision significantly improved. In 2024, the patient developed immune-associated pneumonitis, initially treated with prednisolone. The pulmonary situation became worse. A CT scan confirmed additional metastases in the lung with lymphangiosis carcinomatosis. Due to a poor performance status, no further systemic therapy has been initiated to date. In conclusion, this case highlights the rarity of choroidal metastases in RCC and the challenges of diagnosis. Ophthalmologic examination in RCC patients experiencing sudden vision loss is essential to detect these specific metastases as soon as possible. Comprehensive documentation and awareness of these rare metastatic manifestations are essential to improving diagnostic accuracy and patient outcomes.
Collapse
Affiliation(s)
- Nadja Kührer
- Urology, Medical University of Innsbruck, Innsbruck, AUT
| | - Gertrud Haas
- Ophthalmology, Medical University of Innsbruck, Innsbruck, AUT
| | - Hamed Wafa
- Urology, Medical University of Innsbruck, Innsbruck, AUT
| | | | - Renate Pichler
- Urology, Medical University of Innsbruck, Innsbruck, AUT
| |
Collapse
|
275
|
Kreatsoulas DC, Lonser RR. Spinal cord hemangioblastomas in von Hippel-Lindau disease. Neurooncol Adv 2024; 6:iii66-iii72. [PMID: 39430395 PMCID: PMC11485647 DOI: 10.1093/noajnl/vdad153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024] Open
Abstract
Background von Hippel-Lindau disease (VHL) is an autosomal dominant familial neoplasia syndrome. The most common manifestation of VHL is central nervous system hemangioblastomas. VHL patients will often develop multiple hemangioblastomas along their craniospinal axis over their lifetime. Spinal cord hemangioblastomas account for nearly half of all nervous system hemangioblastomas in VHL. Methods The authors conducted a literature review and summation of available articles on spinal cord hemangioblastomas associated with VHL. Results The embryological origins, epidemiology, natural history, surgical outcomes, nonsurgical treatments, and future directions in spinal cord hemangioblastomas are discussed. Conclusions Hemangioblastomas in VHL are optimally managed with a multidisciplinary approach that includes surgical resection of symptomatic lesions. Novel treatments are gaining traction, but must be studied further for efficacy and safety.
Collapse
Affiliation(s)
- Daniel C Kreatsoulas
- Department of Neurological Surgery, Ohio State University Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
| | - Russell R Lonser
- Department of Neurological Surgery, Ohio State University Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
276
|
Viktor G, Martin B, Mohammad-Reza R, Günter N, Marco S, Anne F, Michael M, Christoph M, Mark-Oliver Z, Anke W, Andreas H, Jochen C, C D, Thomas H, M S, Disorn S, Philipp I. Final Analysis of a Noninterventional Study on Cabozantinib in Patients With Advanced Renal Cell Carcinoma After Prior Checkpoint Inhibitor Therapy of the German Interdisciplinary Working Group on Renal Tumors (IAG-N). Clin Genitourin Cancer 2024; 22:102159. [PMID: 39095295 DOI: 10.1016/j.clgc.2024.102159] [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: 06/18/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Efficacy of treatment after failure of check point inhibitors (ICI) therapy remains ill-defined in metastatic renal cell carcinoma (mRCC). OBJECTIVE To evaluate the safety and effectiveness of cabozantinib after failure of ICI-based therapies. DESIGN, SETTING AND PARTICIPANTS Patients with mRCC who concluded cabozantinib treatment directly after an ICI-based therapy were eligible. Data was collected retrospectively from participating sites in Germany. INTERVENTIONS Cabozantinib was administered as a standard of care. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Adverse events (AE) were reported according to CTCAE v5.0. Objective response rate according to RECIST 1.1 and Progression Free Survival (PFS) were collected from medical records. Descriptive statistics and Kaplan-Meyer-plots were utilized. RESULTS AND LIMITATIONS About 56 eligible patients (71.4% male) with median age of 66 years and clear cell histology in 66.1% (n = 37) were analyzed. 87.5% (n = 49) had ≥ 2 previous lines. IMDC risk was intermediate or poor in 17 patients (30.4%) and missing in 66.1%. 20 patients (35.7%) started with 60 mg. 55.4% (n = 31) required dose reductions, 26.8% (n = 15) treatment delays and 1.8% (n = 1) treatment discontinuation. Partial response was reported in 10.7% (n = 6), stable and progressive disease were reported in 19.6% (n = 11) and in 12.5% (n = 7). 32 patients were not evaluable (57.1%). Median treatment duration was 6.1 months. Treatment related AE were reported in 76.8% (n = 43) and 19.6% (n = 11) had grade 3-5. Fatigue (26.8%), diarrhea (26.8%) and hand-foot-syndrome (25.0%) were the 3 most frequent AEs of any grade and causality. SAE were reported in 21.4% (n = 12), 2 were fatal. Major limitation was the retrospective data capture in our study. CONCLUSIONS Cabozantinib followed directly after ICI-based therapy was safe and feasible. No new safety signals were reported. A lower starting dose was frequently utilized in this real-world cohort, which was associated with a favorable tolerability profile. Our data supports the use of cabozantinib after ICI treatment.
Collapse
Affiliation(s)
- Grünwald Viktor
- Wetsdeutsches Tumorzentrum, Department of Medical Oncology and Department of Urology, University Hospital Essen (AöR), Essen, Germany; Westdeutsches Tumorzentrum Essen, Klinik für Urologie, Universitätsklinikum Essen (AöR), Essen, Germany.
| | - Bögemann Martin
- Klinik für Urologie und Kinderurologie, Universitätsklinikum Münster, Münster, Germany
| | - Rafiyan Mohammad-Reza
- Krankenhaus Nordwest gGmbH, Institut für Klinisch-Onkologische Forschung (IKF), Frankfurt, Germany
| | - Niegisch Günter
- Klinik für Urologie, Konservative Urologische Onkologie, Universitätsklinikum Düsseldorf, Düsseldorf, Germany; Centrum für Integrierte Onkologie (CIO) Düsseldorf, CIO Aachen, Bonn, Köln, Düsseldorf
| | - Schnabel Marco
- Klinik für Urologie der Universität Regensburg am Caritas-Krankenhaus St. Josef, Regensburg, Germany
| | - Flörcken Anne
- Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Deutschland
| | | | | | | | - Wortmann Anke
- Onkologiezentrum Soest-Iserlohn, Medizinisches Versorgungszentrum Kloster Paradiese GbR, Soest-Paradiese, Germany
| | - Hinkel Andreas
- Department for Urology, Franziskus Hospital Bielefeld, Onkologisches Zentrum, Bielefeld, Germany
| | - Casper Jochen
- Klinikum Oldenburg AöR, Universitätsklinik für Innere Medizin - Onkologie und Hämatologie, Oldenburg, Germany
| | - Darr C
- Westdeutsches Tumorzentrum Essen, Klinik für Urologie, Universitätsklinikum Essen (AöR), Essen, Germany
| | - Hilser Thomas
- Wetsdeutsches Tumorzentrum, Department of Medical Oncology and Department of Urology, University Hospital Essen (AöR), Essen, Germany
| | - Schulze M
- Praxis Dr. Schulze, Markkleeberg, Germany
| | - Sookthai Disorn
- Institut für Klinische Krebsforschung IKF GmbH, Krankenhaus Nordwest GmbH, Frankfurt, Germany
| | - Ivanyi Philipp
- Medizinische Hochschule Hannover, Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Hannover, Germany; Comprehensive Cancer Center Hannover; Germany
| |
Collapse
|
277
|
Gunenc D, Issa W, Gerald T, Zhou Q, Zhang S, Ibezue IC, Bhanvadia R, Tachibana I, Brugarolas J, Hammers H, Qin Q, Kapur P, Woldu S, Gaston K, Lotan Y, Cadeddu J, Wang AZ, Margulis V, Zhang T. Pathological Response and Outcomes in Patients With Metastatic Renal Cell Carcinoma (mRCC) Receiving Immunotherapy-Based Therapies and Undergoing Deferred Cytoreductive Nephrectomy (CN). Clin Genitourin Cancer 2024; 22:102177. [PMID: 39218752 DOI: 10.1016/j.clgc.2024.102177] [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: 05/26/2024] [Revised: 07/12/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024]
Abstract
In this study we evaluated outcomes of patients with metastatic renal cell carcinoma who received immunotherapy before surgery. We found that receiving immunotherapy combinations before surgery can offer patients benefits in reducing tumor size and improving disease control. BACKGROUND Immunotherapy (IO) has improved outcomes for patients with metastatic renal cell carcinoma (mRCC). However, the timing of surgical intervention for cytoreductive nephrectomy (CN) is still controversial for this group of patients. PATIENTS AND METHODS We identified patients with mRCC receiving IO-based therapies and undergoing CN. Patients were divided into 2 cohorts: those who underwent upfront CN and those who underwent deferred CN. Pathologic and radiographic features along with clinical outcomes were systematically collected. Comparisons were performed using Chi-square test, paired t-Test or Mann-Whitney-U test. Progression Free survival (PFS) and Overall Survival (OS) were estimated using the Kaplan-Meier method. RESULTS Fifty-one patients with mRCC were included, with a median follow-up of 21 months. 38 (74.5%) patients received IO-based therapies prior to CN, while 13 (25.5%) patients underwent up-front CN. IO-based therapies reduced median tumor size from pretreatment 10 cm to 8.6 cm post-treatment when given prior to CN. IO-TKI had a trend toward higher tumor shrinkage (-2.3 vs -1.2 cm). Pathologic T downstaging occurred in 42% (n=16) of patients, 11% (n=4) of whom had pT0 disease. Thrombus downstaging occurred in 13% (n=6) of patients, all with either partial response (PR) or complete response (CR) in metastases. PFS (HR=0.7, 95% CI 0.29-1.98, p=0.58) and OS (HR 0.4, 95% CI 0.13-1.57, p=0.21) were not statistically significant between 2 cohorts. CONCLUSIONS IO-based therapies, particularly IO-TKIs, resulted in pathologic necrosis and reductions in tumor size prior to deferred CN. PFS and OS were similar for patients who received either upfront IO-based therapy or after CN.
Collapse
Affiliation(s)
- Damla Gunenc
- University of Texas Southwestern, Department of Internal Medicine, Division of Hematology and Oncology, Dallas, TX
| | - Wadih Issa
- University of Texas Southwestern, Department of Internal Medicine, Division of Hematology and Oncology, Dallas, TX
| | - Thomas Gerald
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - Qinhan Zhou
- University of Texas Southwestern, Department of Internal Medicine, Division of Hematology and Oncology, Dallas, TX
| | - Song Zhang
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern, Dallas, TX
| | - I Chidera Ibezue
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - Raj Bhanvadia
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - Isamu Tachibana
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - James Brugarolas
- University of Texas Southwestern, Department of Internal Medicine, Division of Hematology and Oncology, Dallas, TX
| | - Hans Hammers
- University of Texas Southwestern, Department of Internal Medicine, Division of Hematology and Oncology, Dallas, TX
| | - Qian Qin
- University of Texas Southwestern, Department of Internal Medicine, Division of Hematology and Oncology, Dallas, TX
| | - Payal Kapur
- University of Texas Southwestern, Department of Pathology, Dallas, TX
| | - Solomon Woldu
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - Kris Gaston
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - Yair Lotan
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - Jeffrey Cadeddu
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - Andrew Z Wang
- University of Texas Southwestern, Department of Radiation Oncology, Dallas, TX
| | - Vitaly Margulis
- University of Texas Southwestern, Department of Urology, Dallas, TX
| | - Tian Zhang
- University of Texas Southwestern, Department of Internal Medicine, Division of Hematology and Oncology, Dallas, TX.
| |
Collapse
|
278
|
Adhikari A, Sapkota S, Gogia S, Kc O. Changes in the overall survival of patients with metastatic renal cell carcinoma in the era of immune-checkpoint inhibitors. Cancer Epidemiol 2024; 92:102639. [PMID: 39146874 DOI: 10.1016/j.canep.2024.102639] [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: 02/18/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND The advent of immune checkpoint inhibitors (ICI) has brought about a significant transformation in the treatment of immunogenic tumors. On November 23, 2015, the United States Food and Drug Administration approved Nivolumab to treat metastatic renal cell carcinoma (RCC). We aimed to assess potential changes in the survival rates of patients with metastatic RCC at a population level after the approval of Nivolumab. METHODS We used data from the latest version of the Surveillance, Epidemiology, and End Results (SEER) database which encompasses data up to the year 2020. We included patients with age ≥ 20 years who were diagnosed with 'distant' RCC from 2011 through 2020. Based on the approval of Nivolumab, the period from 2011 to 2020 was further grouped into 2011-2015 (pre-ICI era) and 2016-2020 (ICI era). RESULTS The median overall survival (OS) was 8 months in the pre-ICI era compared to 11 months in the ICI era (log-rank test, χ2 = 102.53, p < 0.001). Patients diagnosed with metastatic RCC in the ICI era had a significantly lower risk of dying [Cox proportional Hazard Ratio of 0.77, 95 % CI (0.74-0.80)] compared to patients diagnosed in the pre-ICI era. Additionally, patients under the age of 75 had a lower risk of death compared to those aged 75 years or older. Patients who received chemotherapy (systemic therapy), radiotherapy, or surgery faced a significantly lower risk of mortality. Individuals with metastasis to the brain, bone, liver, or lung had a significantly higher risk of death than those without metastasis to these locations. Marital status also played a role, as married individuals had a significantly lower risk of death compared to those who were divorced, separated, or widowed at the time of diagnosis. Furthermore, income level influenced survival, with patients earning a median annual household income of more than USD 75,000 exhibiting a significantly lower risk of mortality compared to those earning between USD 50,000 and USD 74,000. There was no significant difference in survival observed between non-Hispanic blacks and non-Hispanic whites. CONCLUSION The advent of immune checkpoint inhibitors has led to a substantial improvement in the median overall survival of individuals diagnosed with metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Arjab Adhikari
- Ascension Saint Francis Hospital, 355 Ridge Ave, Evanston, IL 60202, USA.
| | - Supriya Sapkota
- Ascension Saint Francis Hospital, 355 Ridge Ave, Evanston, IL 60202, USA.
| | - Sopiko Gogia
- Ascension Saint Francis Hospital, 355 Ridge Ave, Evanston, IL 60202, USA.
| | - Ojbindra Kc
- Faith Regional Health Services, 2700 W Norfolk Ave, Norfolk, NE 68701, USA.
| |
Collapse
|
279
|
La Vecchia M, Federico M, Aiello D, Zagardo V, Mazzonello A, Testa L, La Paglia L, Bruno T, Fazio I. The Role of Stereotactic Body Radiotherapy (SBRT) in Oligoprogressive Renal Cell Carcinoma (RCC) Treated with ICIs-TKIs: A Retrospective Multicentric Study. J Pers Med 2024; 14:1030. [PMID: 39452537 PMCID: PMC11508468 DOI: 10.3390/jpm14101030] [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: 07/07/2024] [Revised: 08/09/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND This multicentric, retrospective study investigated the use of stereotactic body radiotherapy (SBRT) in patients (pts) with metastatic renal cell carcinoma (mRCC) who experienced oligoprogression during a combination therapy with an immune checkpoint inhibitor (ICI) and a tyrosine-kinase inhibitor (TKI). METHODS We retrospectively evaluated 34 pts affected by oligoprogressive RCC treated with an ICI-TKI combination between January 2020 and December 2023. SBRT was delivered to each site of oligoprogressive metastatic disease. After SBRT, pts were given follow-up clinical evaluations. 6-12-18-month local control (LC) rates and median next-line treatment-free survival (NEST-FS) were the primary endpoints. The secondary endpoints were overall response rate (ORR), clinical benefits and safety. RESULTS After a median follow-up of 24 months, 6-12-18-month LC rates were 100%, 71% and 43%, respectively, and the median NEST-FS was 20 months. ORR was 90%, while clinical benefit was 100%. No > G2 adverse events related to SBRT were recorded. CONCLUSIONS In our study, SBRT for oligoprogressive mRCC turned out to be a safe and useful treatment which was able to preserve current treatment. Further prospective studies are necessary to explore the effects of the ICIs-TKIs combination and SBRT upon oligoprogressive sites in mRCC.
Collapse
Affiliation(s)
- Maria La Vecchia
- Unità Operativa di Radioterapia Oncologica, Casa di Cura Macchiarella, 90138 Palermo, Italy; (M.L.V.); (D.A.); (A.M.); (L.T.); (L.L.P.); (T.B.); (I.F.)
| | - Manuela Federico
- Unità Operativa di Radioterapia Oncologica, Casa di Cura Macchiarella, 90138 Palermo, Italy; (M.L.V.); (D.A.); (A.M.); (L.T.); (L.L.P.); (T.B.); (I.F.)
| | - Dario Aiello
- Unità Operativa di Radioterapia Oncologica, Casa di Cura Macchiarella, 90138 Palermo, Italy; (M.L.V.); (D.A.); (A.M.); (L.T.); (L.L.P.); (T.B.); (I.F.)
| | - Valentina Zagardo
- Unità Operativa di Radioterapia Oncologica, Rem Radioterapia srl, 95029 Viagrande, Italy;
| | - Antonella Mazzonello
- Unità Operativa di Radioterapia Oncologica, Casa di Cura Macchiarella, 90138 Palermo, Italy; (M.L.V.); (D.A.); (A.M.); (L.T.); (L.L.P.); (T.B.); (I.F.)
| | - Lorella Testa
- Unità Operativa di Radioterapia Oncologica, Casa di Cura Macchiarella, 90138 Palermo, Italy; (M.L.V.); (D.A.); (A.M.); (L.T.); (L.L.P.); (T.B.); (I.F.)
| | - Leonarda La Paglia
- Unità Operativa di Radioterapia Oncologica, Casa di Cura Macchiarella, 90138 Palermo, Italy; (M.L.V.); (D.A.); (A.M.); (L.T.); (L.L.P.); (T.B.); (I.F.)
| | - Tiziana Bruno
- Unità Operativa di Radioterapia Oncologica, Casa di Cura Macchiarella, 90138 Palermo, Italy; (M.L.V.); (D.A.); (A.M.); (L.T.); (L.L.P.); (T.B.); (I.F.)
| | - Ivan Fazio
- Unità Operativa di Radioterapia Oncologica, Casa di Cura Macchiarella, 90138 Palermo, Italy; (M.L.V.); (D.A.); (A.M.); (L.T.); (L.L.P.); (T.B.); (I.F.)
| |
Collapse
|
280
|
Chen H, Liu H, Zhang X, Wang S, Liu C, An K, Liu R, Tian X. Diversified applications of hepatocellular carcinoma medications: molecular-targeted, immunotherapeutic, and combined approaches. Front Pharmacol 2024; 15:1422033. [PMID: 39399471 PMCID: PMC11467865 DOI: 10.3389/fphar.2024.1422033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the primary forms of liver cancer and is currently the sixth most prevalent malignancy worldwide. In addition to surgical interventions, effective drug treatment is essential for treating HCC. With an increasing number of therapeutic drugs for liver cancer undergoing clinical studies, the therapeutic strategies for advanced HCC are more diverse than ever, leading to improved prospects for HCC patients. Molecular targeted drugs and immunotherapies have become crucial treatment options for HCC. Treatment programs include single-agent molecular-targeted drugs, immunotherapies, combinations of immunotherapies with molecular-targeted drugs, and dual immune checkpoint inhibitors. However, further exploration is necessary to determine the optimal pharmacological treatment regimens, and the development of new effective drugs is urgently needed. This review provides an overview of the current globally approved drugs for liver cancer, as well as the latest advances in ongoing clinical research and drug therapies. Additionally, the review offers an outlook and discussion on the prospects for the development of drug therapy approaches for HCC.
Collapse
Affiliation(s)
- Haoyang Chen
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Huihui Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Xiaowei Zhang
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Suhua Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Chunxia Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Ke An
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Ruijuan Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| | - Xin Tian
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou, China
| |
Collapse
|
281
|
Compagno S, Casadio C, Galvani L, Rosellini M, Marchetti A, Tassinari E, Piazza P, Mottaran A, Santoni M, Schiavina R, Massari F, Mollica V. Novel Immune Checkpoint Inhibitor Targets in Advanced or Metastatic Renal Cell Carcinoma: State of the Art and Future Perspectives. J Clin Med 2024; 13:5738. [PMID: 39407796 PMCID: PMC11476392 DOI: 10.3390/jcm13195738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have become the cornerstone of treatment in renal cell carcinoma (RCC), for both metastatic disease and in an adjuvant setting. However, an adaptive resistance from cancer cells may arise during ICI treatment, therefore many studies are focusing on additional immune checkpoint inhibitor pathways. Promising targets of immunotherapeutic agents under investigation include T cell immunoglobulin and ITIM domain (TIGIT), immunoglobulin-like transcript 4 (ILT4), lymphocyte activation gene-3 (LAG-3), vaccines, T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), and chimeric antigen receptor (CAR) T cells. In this review of the literature, we recollect the current knowledge of the novel treatment strategies in the field of immunotherapy that are being investigated in RCC and analyze their mechanism of action, their activity and the clinical studies that are currently underway.
Collapse
Affiliation(s)
- Samuele Compagno
- Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.C.); (C.C.); (L.G.); (M.R.); (A.M.); (E.T.); (F.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
| | - Chiara Casadio
- Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.C.); (C.C.); (L.G.); (M.R.); (A.M.); (E.T.); (F.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
| | - Linda Galvani
- Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.C.); (C.C.); (L.G.); (M.R.); (A.M.); (E.T.); (F.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
| | - Matteo Rosellini
- Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.C.); (C.C.); (L.G.); (M.R.); (A.M.); (E.T.); (F.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
| | - Andrea Marchetti
- Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.C.); (C.C.); (L.G.); (M.R.); (A.M.); (E.T.); (F.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
| | - Elisa Tassinari
- Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.C.); (C.C.); (L.G.); (M.R.); (A.M.); (E.T.); (F.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
| | - Pietro Piazza
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
- Division of Urology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Angelo Mottaran
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
- Division of Urology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Riccardo Schiavina
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
- Division of Urology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.C.); (C.C.); (L.G.); (M.R.); (A.M.); (E.T.); (F.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (P.P.); (A.M.); (R.S.)
| | - Veronica Mollica
- Medical Oncology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.C.); (C.C.); (L.G.); (M.R.); (A.M.); (E.T.); (F.M.)
| |
Collapse
|
282
|
Xiong B, Liu W, Liu Y, Chen T, Lin A, Song J, Qu L, Luo P, Jiang A, Wang L. A Multi-Omics Prognostic Model Capturing Tumor Stemness and the Immune Microenvironment in Clear Cell Renal Cell Carcinoma. Biomedicines 2024; 12:2171. [PMID: 39457484 PMCID: PMC11504857 DOI: 10.3390/biomedicines12102171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Cancer stem-like cells (CSCs), a distinct subset recognized for their stem cell-like abilities, are intimately linked to the resistance to radiotherapy, metastatic behaviors, and self-renewal capacities in tumors. Despite their relevance, the definitive traits and importance of CSCs in the realm of oncology are still not fully comprehended, particularly in the context of clear cell renal cell carcinoma (ccRCC). A comprehensive understanding of these CSCs' properties in relation to stemness, and their impact on the efficacy of treatment and resistance to medication, is of paramount importance. Methods: In a meticulous research effort, we have identified new molecular categories designated as CRCS1 and CRCS2 through the application of an unsupervised clustering algorithm. The analysis of these subtypes included a comprehensive examination of the tumor immune environment, patterns of metabolic activity, progression of the disease, and its response to immunotherapy. In addition, we have delved into understanding these subtypes' distinctive clinical presentations, the landscape of their genomic alterations, and the likelihood of their response to various pharmacological interventions. Proceeding from these insights, prognostic models were developed that could potentially forecast the outcomes for patients with ccRCC, as well as inform strategies for the surveillance of recurrence after treatment and the handling of drug-resistant scenarios. Results: Compared with CRCS1, CRCS2 patients had a lower clinical stage/grading and a better prognosis. The CRCS2 subtype was in a hypoxic state and was characterized by suppression and exclusion of immune function, which was sensitive to gefitinib, erlotinib, and saracatinib. The constructed prognostic risk model performed well in both training and validation cohorts, helping to identify patients who may benefit from specific treatments or who are at risk of recurrence and drug resistance. A novel therapeutic target, SAA2, regulating neutrophil and fibroblast infiltration, and, thus promoting ccRCC progression, was identified. Conclusions: Our findings highlight the key role of CSCs in shaping the ccRCC tumor microenvironment, crucial for therapy research and clinical guidance. Recognizing tumor stemness helps to predict treatment efficacy, recurrence, and drug resistance, informing treatment strategies and enhancing ccRCC patient outcomes.
Collapse
Affiliation(s)
- Beibei Xiong
- Department of Oncology, The First People’s Hospital of Shuangliu District, Chengdu 610200, China;
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Ying Liu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Tong Chen
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; (A.L.); (P.L.)
| | - Jiaao Song
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China;
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; (A.L.); (P.L.)
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| |
Collapse
|
283
|
Abstract
Importance Renal cell carcinoma (RCC) is a common malignancy, with an estimated 434 840 incident cases worldwide in 2022. In the US, it is the sixth most common cancer among males and ninth among females. Observations Clear cell RCC is the most common histologic subtype (75%-80% of cases) and is characterized by inactivation of the von Hippel Lindau (VHL) tumor suppressor gene. Many patients (37%-61%) are diagnosed with RCC incidentally on an abdominal imaging study such as ultrasound or computed tomographic scan, and 70% of patients have stage I RCC at diagnosis. Although its incidence has increased approximately 1% per year from 2015 through 2019, the mortality rate of RCC has declined about 2% per year in the US from 2016 through 2020. Patients with a solid renal mass or complex cystic renal mass should be referred to urology. Treatment options for RCC confined to the kidney include surgical resection with partial or radical nephrectomy, ablative techniques (eg, cryoablation, radiofrequency ablation, radiation), or active surveillance for some patients (especially those with renal masses <2 cm). For patients with renal masses less than 4 cm in size (48% of patients), partial nephrectomy can result in a 5-year cancer-specific survival of more than 94%. For advanced or metastatic RCC, combinations of immune checkpoint inhibitors or the combination of immune checkpoint inhibitors with tyrosine kinase inhibitors are associated with tumor response of 42% to 71%, with a median overall survival of 46 to 56 months. Conclusions and Relevance RCC is a common malignancy that is often diagnosed incidentally on an abdominal imaging study. Seventy percent of patients are diagnosed with stage I RCC and 11% of patients with stage IV. First-line treatments for early-stage RCC are partial or radical nephrectomy, which can result in 5-year cancer-specific survival of more than 94%, ablative techniques, or active surveillance. New treatment options for patients with metastatic RCC include immune checkpoint inhibitors and tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Tracy L Rose
- Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
| | - William Y Kim
- Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
- Department of Genetics, University of North Carolina at Chapel Hill
- Department of Pharmacology, University of North Carolina at Chapel Hill
| |
Collapse
|
284
|
Al-Mansour MM, Aga SS, Alharbi HA, Alsulami MN, Fallatah HA, Albedaiwi TB, Anbari LK, Surrati TR, Algethami AA, Althubaiti A, Alfayea TM, Alolayan A. Real-World Survival Outcomes of First-Line Therapies in Patients with Metastatic Clear Cell Renal Cell Carcinoma: A Retrospective Analysis from Two Centres in Saudi Arabia. Cancers (Basel) 2024; 16:3234. [PMID: 39335205 PMCID: PMC11430578 DOI: 10.3390/cancers16183234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Metastatic renal cell carcinoma (mRCC) represents a challenging condition characterised by poor prognosis and limited response to chemoradiotherapy. In this retrospective study, we compared the survival outcomes of first-line ICI regimens versus single-agent TKIs in patients with mRCC from two centres in Saudi Arabia. Methods: This study included 84 patients diagnosed with clear cell mRCC between January 2016 and December 2023. Patients were grouped based on treatment regimens. Progression-free survival (PFS) and overall survival (OS) were analysed using Kaplan-Meier curves and Cox proportional hazards regression. Results: The median first-line PFS was 9.7 months (95% CI: 5.3-14.1) for the overall cohort, with no significant difference between the single-agent tyrosine kinase inhibitor (TKI) group (9.4 months; 95% CI: 6.4-12.4), combination ICI group (9.0 months; 95% CI: 0.0-24.9), and single-agent ICI group (21.2 months; 95% CI: 2.6-39.8; p = 0.591). The median OS for the overall cohort was 42.0 months (95% CI: 14.9-69.2), with the single-agent TKI group having a median OS of 33.3 months (95% CI: 0.0-71.7), the combination ICI group, 42.0 months (95% CI: 0.06-84.0), and the single-agent ICI group, 23.0 months (95% CI: 19.2-26.7; p = 0.73). In comparison, the ICI-based combination therapy group exhibited a higher ORR of 41.0% (95% CI: 26.3-57.8%), while the single-agent ICI group had an ORR of 20.0% (95% CI: 3.5-55.8%). Cox regression identified liver metastasis as a significant independent predictor of PFS (HR = 1.8, p = 0.043), while a lower Karnofsky Performance Status was a significant independent predictor of OS (HR = 3.5, p < 0.001). Conclusions: In real-world practice from Saudi Arabia, first-line, single-agent ICI therapy offers promising anti-tumour activity and non-inferior survival outcomes compared to standard ICI-based combinations and single-agent TKIs.
Collapse
Affiliation(s)
- Mubarak M Al-Mansour
- Adult Medical Oncology, Princess Noorah Oncology Centre, Ministry of National Guard Health Affairs-Western Region (MNGHA-WR), King Abdullah International Medical Research Centre (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
- Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Syed Sameer Aga
- Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Hanin A Alharbi
- Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Maria N Alsulami
- Department of Clinical Pharmacy, Pharmaceutical Care Services, Ministry of National Guard Health Affairs-Western Region (MNGHA-WR), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Halah A Fallatah
- Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Tarfah B Albedaiwi
- Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Lujain K Anbari
- Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Taleen R Surrati
- Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Ashwag A Algethami
- Department of Clinical Pharmacy, Pharmaceutical Care Services, Ministry of National Guard Health Affairs-Western Region (MNGHA-WR), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Alaa Althubaiti
- Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Centre (KAIMRC), King Abdulaziz Medical City, Jeddah 21423, Saudi Arabia
| | - Turki M Alfayea
- Adult Medical Oncology Department, Ministry of National Guard Health Affairs-Central Region (MNGHA-CR), King Abdullah International Medical Research Centre (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, Riyadh 11481, Saudi Arabia
| | - Ashwaq Alolayan
- Adult Medical Oncology Department, Ministry of National Guard Health Affairs-Central Region (MNGHA-CR), King Abdullah International Medical Research Centre (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, Riyadh 11481, Saudi Arabia
| |
Collapse
|
285
|
Roy S, Raychaudhuri S, Biswas B, Dabkara D, Bhattacherjee A, Ganguly S, Ghosh J, Patel YS, Pal S, Karmakar J, Mitra A, Gupta S. Pattern of care and treatment outcomes of metastatic non-clear cell kidney cancer: a single centre experience from India. Ecancermedicalscience 2024; 18:1775. [PMID: 39430082 PMCID: PMC11489090 DOI: 10.3332/ecancer.2024.1775] [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: 03/04/2024] [Indexed: 10/22/2024] Open
Abstract
Background Non-clear-cell renal cell carcinoma (nccRCC) refers to a rare diverse heterogeneous group of tumours; usually treated with immune check point inhibitors and or tyrosine kinase inhibitors (TKIs). Prospective large-scale data from Asian countries is limited. Methods This is a retrospective study of patients with metastatic nccRCC treated at Tata Medical Centre, Kolkata, India, from 2012 to 2022. Demographic profiles, histologic subtypes, treatment details, response to therapy (by response evaluation criteria in solid tumours (RECIST v1.1)) and survival status were captured from electronic medical records (EMRs) of hospitals up till May 2023. Kaplan Meier methods were estimated to assess progression-free survival (PFS) and overall survival (OS). Results A total of 89 consecutive patients were screened for this study, 24 were excluded due to inadequate data in EMR. 65 patients were included in the final analysis, with a median age at diagnosis of 59 years (range 20-84) of which 81% were male. Histologic subtypes comprised of 43% papillary, 31% clear cell with mixed histology, 3% sarcomatoid and 23% others including chromophobe, mucinous-tubular, spindle cell, oncocytic, medullary, poorly differentiated and rhabdoid). The most common site of metastasis was the lung 62% (n = 40) followed by non-regional nodes 32%, bone 26% and liver 14%. 15% patients presented with haematuria and 62% underwent nephrectomy prior to systemic therapy. The majority received pazopanib 46% (n = 30), chemotherapy 20% (n = 13) including bevacizumab plus erlotinib, sunitinib 15% (n = 10) or cabozantinib 14% (n = 9). Only 3(5%) patients received nivolumab plus cabozantinib combination. Response to treatment showed complete response in 1.5%, partial response in 20%, stable disease in 51% and progressive disease in 23% as per RECIST v1.1. 17 patients required dose reduction and interruption due to adverse effects and 33% (n = 22) received second-line therapy with nivolumab 18% (n = 4), axitinib and everolimus among others. After a median follow up of 44 months, the median PFS was 13 months (95%CI 7.2-18.9) and the median OS was 17 months (95%CI 12.1-22.1) for the entire cohort. Conclusion The overall response and survival for metastatic nccRCC was relatively better in comparison with published data, despite the limited number of patients treated with ICIs due to cost and access barriers.
Collapse
Affiliation(s)
- Somnath Roy
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Sreejata Raychaudhuri
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Bivas Biswas
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Deepak Dabkara
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Arnab Bhattacherjee
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Sandip Ganguly
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Joydeep Ghosh
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Yesha Sandipbhai Patel
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Souhita Pal
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Jagriti Karmakar
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Anindita Mitra
- Department of Medical Oncology, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| | - Sujoy Gupta
- Uro-Surgery, Tata Medical Center, 14 MAR (EW), New Town, Rajarhat, Kolkata 700160, West Bengal, India
| |
Collapse
|
286
|
Omri L, Naigeon M, Flippot R, Gavira-Díaz J, Poveda-Ferriols J, Nguyen D, Abdi C, Arroyo-Salgado A, Chaput N, de Velasco G, Albigès L, Carril-Ajuria L. Blood-based circulating biomarkers for prediction of immune-checkpoint inhibitors efficacy in renal cell carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1199-1222. [PMID: 39465007 PMCID: PMC11502076 DOI: 10.37349/etat.2024.00271] [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: 06/17/2024] [Accepted: 08/09/2024] [Indexed: 10/29/2024] Open
Abstract
Immune checkpoint inhibitors (ICI)-based combinations have become the standard first-line treatment for advanced clear cell renal cell carcinoma (ccRCC). Despite significant improvements in survival and the achievement of sustained long-term responses, a subset of patients remains refractory to ICI, and most will eventually develop resistance. Thus, identifying predictive biomarkers for ICI efficacy and resistance is essential for optimizing therapeutic strategies. Up to now, tissue-based biomarkers have not been successful as predictive biomarkers in RCC. Circulating blood-based biomarkers offer a promising alternative. These biomarkers, including circulating immune cells, soluble factors, tumor-derived markers, and those based on metabolomics, are less invasive, offer reproducibility over time, and provide a comprehensive assessment of tumor biology and patient immune status, as well as allow dynamic monitoring during treatment. This review aims to evaluate the current evidence on the different candidate circulating biomarkers being investigated for their potential to predict ICI efficacy in RCC patients.
Collapse
Affiliation(s)
- Loubna Omri
- Department of Medical Oncology, National Institute of Oncology, Rabat X4FH+66, Morocco
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
| | - Marie Naigeon
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94805 Villejuif, France
- Paris-Saclay University, School of Pharmacy, 91190 Orsay, France
| | - Ronan Flippot
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94805 Villejuif, France
- Medical Oncology Department, Institut Gustave Roussy, 94805 Villejuif, France
| | - Javier Gavira-Díaz
- Medical Oncology Department, Institut Gustave Roussy, 94805 Villejuif, France
| | - Jesus Poveda-Ferriols
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
- Medical Oncology Department, Centre Hospitalier Universitaire Saint-Pierre, 1000 Brussels, Belgium
| | - Dan Nguyen
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
| | - Chaimae Abdi
- Department of Medical Oncology, National Institute of Oncology, Rabat X4FH+66, Morocco
| | - Alvaro Arroyo-Salgado
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94805 Villejuif, France
| | - Guillermo de Velasco
- Medical Oncology Department, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Laurence Albigès
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94805 Villejuif, France
- Medical Oncology Department, Institut Gustave Roussy, 94805 Villejuif, France
| | - Lucía Carril-Ajuria
- Medical Oncology Department, Centre Hospitalier Universitaire Brugmann, 1020 Brussels, Belgium
- Medical Oncology Department, Centre Hospitalier Universitaire Saint-Pierre, 1000 Brussels, Belgium
| |
Collapse
|
287
|
Leung DKW, Siu BWH, Teoh JYC. Adjuvant treatment for renal cell carcinoma: current status and future. Curr Opin Urol 2024:00042307-990000000-00192. [PMID: 39298572 DOI: 10.1097/mou.0000000000001229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
PURPOSE OF REVIEW Renal cell carcinoma (RCC) is resistant to chemotherapy. Adjuvant interferon and tyrosine kinase inhibitors were ineffective. Immune checkpoint inhibitors (ICIs), however, have shed new hope in this setting. In the current review, updated evidence of adjuvant therapy in RCC is summarized. RECENT FINDINGS KEYNOTE-564 demonstrated survival benefits of adjuvant Pembrolizumab in RCC. EAU guidelines now recommend adjuvant pembrolizumab to ccRCC patients at an increased risk of recurrence, as defined in the study. At a median follow-up of 24 months, the disease-free survival (DFS) was significantly longer for the Pembrolizumab group than placebo group [DFS 77.3 vs. 68.1%; hazard ratio for recurrence or death, 0.68; 95% confidence interval (95% CI), 0.53-0.87; P = 0.002]. From its updated analysis, at median follow up of 57.2 months, overall survival (OS) benefit of Pembrolizumab was demonstrated (hazard ratio for death, 0.62; 95% CI, 0.44-0.87; P = 0.005). A number of other adjuvant ICI trials have though been negative. SUMMARY Pembrolizumab is currently the only adjuvant therapy for RCC showing survival benefits, amid a number of negative trials on adjuvant immunotherapy. Currently, there is no role for adjuvant tyrosine-kinase inhibitors and radiotherapy for RCC. Meanwhile, a multidisciplinary approach and shared decision-making should be adopted.
Collapse
Affiliation(s)
- David K W Leung
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital
| | - Brian W H Siu
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital
| | - Jeremy Y C Teoh
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Urology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
288
|
Derks SHAE, van der Meer EL, Joosse A, de Jonge MJA, Slagter C, Schouten JW, Hoop EOD, Smits M, van den Bent MJ, Jongen JLM, van der Veldt AAM. The development of brain metastases in patients with different therapeutic strategies for metastatic renal cell cancer. Int J Cancer 2024; 155:1045-1052. [PMID: 38703351 DOI: 10.1002/ijc.34984] [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: 01/09/2024] [Revised: 02/27/2024] [Accepted: 04/09/2024] [Indexed: 05/06/2024]
Abstract
A diagnosis of brain metastasis (BM) significantly affects quality of life in patients with metastatic renal cell cancer (mRCC). Although systemic treatments have shown efficacy in mRCC, active surveillance (AS) is still commonly used in clinical practice. In this single-center cohort study, we assessed the impact of different initial treatment strategies for metastatic RCC (mRCC) on the development of BM. All consecutive patients diagnosed with mRCC between 2011 and 2022 were included at the Erasmus MC Cancer Institute, the Netherlands, and a subgroup of patients with BM was selected. In total, 381 patients with mRCC (ECM, BM, or both) were identified. Forty-six patients had BM of whom 39 had metachronous BM (diagnosed ≥1 month after ECM). Twenty-five (64.1%) of these 39 patients with metachronous BM had received prior systemic treatment for ECM and 14 (35.9%) patients were treatment naive at BM diagnosis. The median BM-free survival since ECM diagnosis was significantly longer (p = .02) in previously treated patients (29.0 [IQR 12.6-57.0] months) compared to treatment naive patients (6.8 [IQR 1.0-7.0] months). In conclusion, patients with mRCC who received systemic treatment for ECM prior to BM diagnosis had a longer BM-free survival as compared to treatment naïve patients. These results emphasize the need for careful evaluation of treatment strategies, and especially AS, for patients with mRCC.
Collapse
Affiliation(s)
- Sophie H A E Derks
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Edgar L van der Meer
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Maja J A de Jonge
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Cleo Slagter
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Joost W Schouten
- Department of Neurosurgery, Erasmus MC, Rotterdam, The Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marion Smits
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Martin J van den Bent
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Joost L M Jongen
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
289
|
Tsimafeyeu I. Sunitinib in Patients with Metastatic Renal Cell Carcinoma with Favorable Risk: Be Aware of PD-L1 Expression. Med Sci (Basel) 2024; 12:48. [PMID: 39311161 PMCID: PMC11417775 DOI: 10.3390/medsci12030048] [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: 08/15/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024] Open
Abstract
The treatment landscape for metastatic renal cell carcinoma (RCC) has advanced significantly with first-line immunotargeted therapy combinations. However, no statistically significant differences were observed in the cohort of patients with favorable risk and some oncologists continue to use sunitinib in these patients. PD-L1 expression has emerged as a negative prognostic factor in RCC, particularly in sunitinib-treated patients, where higher PD-L1 levels are linked to worse outcomes. This article discusses the potential risks associated with the use of sunitinib in PD-L1-positive patients.
Collapse
Affiliation(s)
- Ilya Tsimafeyeu
- Bureau for Cancer Research-BUCARE, 526 W 158th Str., New York, NY 10032, USA
| |
Collapse
|
290
|
Ju Y, Xu D, Liao MM, Sun Y, Bao WD, Yao F, Ma L. Barriers and opportunities in pancreatic cancer immunotherapy. NPJ Precis Oncol 2024; 8:199. [PMID: 39266715 PMCID: PMC11393360 DOI: 10.1038/s41698-024-00681-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/27/2024] [Indexed: 09/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a fatal clinical challenge characterized by a dismal 5-year overall survival rate, primarily due to the lack of early diagnosis and limited therapeutic efficacy. Immunotherapy, a proven success in multiple cancers, has yet to demonstrate significant benefits in PDAC. Recent studies have revealed the immunosuppressive characteristics of the PDAC tumor microenvironment (TME), including immune cells with suppressive properties, desmoplastic stroma, microbiome influences, and PDAC-specific signaling pathways. In this article, we review recent advances in understanding the immunosuppressive TME of PDAC, TME differences among various mouse models of pancreatic cancer, and the mechanisms underlying resistance to immunotherapeutic interventions. Furthermore, we discuss the potential of targeting cancer cell-intrinsic pathways and TME components to sensitize PDAC to immune therapies, providing insights into strategies and future perspectives to break through the barriers in improving pancreatic cancer treatment.
Collapse
Affiliation(s)
- Yixin Ju
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, Guangdong, 518000, China
| | - Dongzhi Xu
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, Guangdong, 518000, China
| | - Miao-Miao Liao
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wen-Dai Bao
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Fan Yao
- Hubei Hongshan Laboratory, College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, Guangdong, 518000, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong, 518000, China.
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
291
|
Anderson AC, Ho J, Hall ET, Hannan R, Liao JJ, Louie AV, Ma TM, Psutka SP, Rengan R, Siva S, Swaminath A, Tachiki L, Tang C, Teh BS, Tsai J, Tykodi SS, Weg E, Zaorsky NG, Lo SS. Focal therapy for oligometastatic and oligoprogressive renal cell carcinoma: a narrative review. Future Oncol 2024; 20:2573-2588. [PMID: 39258792 PMCID: PMC11534104 DOI: 10.1080/14796694.2024.2389769] [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: 05/22/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Metastatic renal cell carcinoma (RCC) can present with oligometastatic disease and/or develop oligoprogression following systemic therapy. Cytoreductive and focal metastasis-directed therapy options include resection, stereotactic ablative radiation and thermal ablation. Aggressive focal therapy may allow delay in initiation of or modification to systemic therapy and improve clinical outcomes. In this narrative review we synthesize current practice guidelines and prospective data on focal therapy management options and highlight future research. Patient selection and the choice of focal treatment techniques are controversial due to limited and heterogeneous data and patients may benefit from multidisciplinary evaluation. Prospective comparative trials with clearly defined inclusion criteria and relevant end points are needed to clarify the risks and benefits of different approaches.
Collapse
Affiliation(s)
- August C Anderson
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Joel Ho
- Pfizer Inc., Bothell, WA 98011, USA
| | - Evan T Hall
- University of Washington, Division of Hematology & Oncology, Seattle, WA 98195,USA
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA 98195, USA
| | - Raquibul Hannan
- The University of Texas Southwestern Medical Center, Radiation Oncology, Dallas, TX 75235, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Kidney Cancer Program, Dallas, TX75235, USA
| | - Jay J Liao
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Alexander V Louie
- Sunnybrook Health Sciences Centre, Department of Radiation Oncology, Toronto, ON, M4N 3M5, Canada
| | - Ting Martin Ma
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Sarah P Psutka
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA 98195, USA
- University of Washington, Department of Urology, Seattle, WA 98195, USA
| | - Ramesh Rengan
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Shankar Siva
- Peter MacCallum Cancer Centre, Division of Radiation Oncology & Cancer Imaging, Melbourne, VIC, 3052, Australia
- The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, VIC, 3052, Australia
| | - Anand Swaminath
- Juravinski Cancer Centre, Radiation Therapy, Hamilton, ON, L8V 5C2, Canada
- McMaster University, Division of Radiation Oncology, Hamilton, ON,L8S 4L8,Canada
| | - Lisa Tachiki
- University of Washington, Division of Hematology & Oncology, Seattle, WA 98195,USA
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA 98195, USA
| | - Chad Tang
- The University of Texas MD Anderson Cancer Center, Genitourinary Radiation Oncology, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center, Investigational Cancer Therapeutics, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center, Translational Molecular Pathology, Houston, TX 77030, USA
| | - Bin Sing Teh
- Houston Methodist Hospital, Radiation Oncology, Houston, TX 77030, USA
| | - Joseph Tsai
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Scott S Tykodi
- University of Washington, Division of Hematology & Oncology, Seattle, WA 98195,USA
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA 98195, USA
| | - Emily Weg
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Nicholas G Zaorsky
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Department of Radiation Oncology, Cleveland, OH 44106, USA
| | - Simon S Lo
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| |
Collapse
|
292
|
Barisic S, Brahmbhatt EM, Cherkasova E, Spear TT, Savani U, Pierre S, Scurti GM, Chen L, Igboko M, Nadal R, Zeng G, Parry G, Stroncek DF, Highfill S, Dalheim AV, Reger R, Nishimura MI, Childs RW. Regression of renal cell carcinoma by T cell receptor-engineered T cells targeting a human endogenous retrovirus. J Immunother Cancer 2024; 12:e009147. [PMID: 39266213 PMCID: PMC11409391 DOI: 10.1136/jitc-2024-009147] [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] [Accepted: 07/31/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND We discovered a novel human endogenous retrovirus (CT-RCC HERV-E) that was selectively expressed in most clear cell renal cell carcinomas (ccRCC) and served as a source of antigens for T cell-mediated killing. Here, we described the cloning of a novel T cell receptor (TCR) targeting a CT-RCC HERV-E-derived antigen specific to ccRCC and characterized antitumor activity of HERV-E TCR-transduced T cells (HERV-E T cells). METHODS We isolated a CD8+ T cell clone from a patient with immune-mediated regression of ccRCC post-allogeneic stem cell transplant that recognized the CT-RCC-1 HERV-E-derived peptide in an HLA-A11-restricted manner. We used 5'Rapid Amplification of cDNA Ends (RACE) to clone the full length HERV-E TCR and generated retrovirus encoding this TCR for transduction of T cells. We characterized HERV-E T cells for phenotype and function in vitro and in a murine xenograft model. Lastly, we implemented a good manufacturing practice-compliant method for scalable production of HERV-E T cells. RESULTS The HLA-A11-restricted HERV-E-reactive TCR exhibited a CD8-dependent phenotype and demonstrated specific recognition of the CT-RCC-1 peptide. CD8+ T cells modified to express HERV-E TCR displayed potent antitumor activity against HLA-A11+ ccRCC cells expressing CT-RCC HERV-E compared with unmodified T cells. Killing by HERV-E T cells was lost when cocultured against HERV-E knockout ccRCC cells. HERV-E T cells induced regression of established ccRCC tumors in a murine model and improved survival of tumor-bearing mice. Large-scale production of HERV-E T cells under good manufacturing practice conditions generated from healthy donors retained specific antigen recognition and cytotoxicity against ccRCC. CONCLUSIONS This is the first report showing that human ccRCC cells can be selectively recognized and killed by TCR-engineered T cells targeting a HERV-derived antigen. These preclinical findings provided the foundation for evaluating HERV-E TCR-transduced T cell infusions in patients with metastatic ccRCC in a clinical trial (NCT03354390).
Collapse
Affiliation(s)
- Stefan Barisic
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Elena Cherkasova
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Timothy T Spear
- Department of Surgery, Loyola University Chicago, Chicago, Illinois, USA
| | - Ujjawal Savani
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephanie Pierre
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gina M Scurti
- Department of Surgery, Loyola University Chicago, Chicago, Illinois, USA
| | - Long Chen
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Muna Igboko
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Rosa Nadal
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gang Zeng
- T-Cure BioScience, Sherman Oaks, California, USA
| | - Gordon Parry
- T-Cure BioScience, Sherman Oaks, California, USA
| | - David F Stroncek
- Center for Cellular Engineering, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Steven Highfill
- Center for Cellular Engineering, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Annika V Dalheim
- Department of Surgery, Loyola University Chicago, Chicago, Illinois, USA
| | - Robert Reger
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Richard W Childs
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
293
|
Yamamoto Y, Tomoto K, Teshigawara A, Ishii T, Hasegawa Y, Akasaki Y, Murayama Y, Tanaka T. Significance and Priority of Surgical Resection as Therapeutic Strategy Based on Clinical Characteristics of Brain Metastases from Renal Cell Carcinoma. World Neurosurg 2024:S1878-8750(24)01535-3. [PMID: 39243967 DOI: 10.1016/j.wneu.2024.08.166] [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: 08/25/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
OBJECTIVE To clarify a rational surgical priority, clinical characteristics were compared between brain metastases (BM) from renal cell carcinoma (RCC) and other cancers. METHODS We reviewed 425 consecutive patients with BM who underwent treatments including surgery between January 2014 and December 2022. Primary cancers included lung (n = 220), breast (n = 46), digestive (n = 65), RCC (n = 25), and others (n = 69). Tumor volume (T), edema volume (E), and edema volume/tumor volume ratio (E/T ratio) were compared between RCC and other primary cancers. Cutoff T values for identifying both symptomatic tumors and tumors suitable for surgery were determined by receiver operating characteristic curves. Factors including E/T ratio, age, Karnofsky Performance Scale score, and tumor characteristics were statistically analyzed. RESULTS Cutoff values of T and E to determine surgical suitability were 4.973 cm3 (sensitivity, 0.848; specificity, 0.74) and 23.088 cm3 (sensitivity, 0.894; specificity, 0.623), respectively. E/T ratio was significantly higher for RCC than for other cancers (P < 0.01). These results remained consistent after propensity score matching. RCC tended to show a significantly lower frequency of posterior fossa tumor (16%, P < 0.01) and higher rates of single lesions (72%, P = 0.03) and intratumoral hemorrhage (24%, P = 0.02). Subgroup analysis limited to surgical cases showed that E was consistent across tumors, T tended to be smaller, and E/T ratio was significantly higher in RCC. CONCLUSIONS Generally, symptomatic BM were indicated for surgery. BM from RCC were characteristically single, low-volume lesions with expanding edema and intratumoral hemorrhage, causing symptoms. These results suggest that surgery should be a high priority for BM from RCC.
Collapse
Affiliation(s)
- Yohei Yamamoto
- Department of Neurosurgery, The Jikei University School of Medicine Daisan Hospital Tokyo, Tokyo, Japan; Department of Neurosurgery, The Jikei University School of Medicine Kashiwa Hospital, Chiba, Japan
| | - Kyoichi Tomoto
- Department of Neurosurgery, The Jikei University School of Medicine Kashiwa Hospital, Chiba, Japan; Department of Neurosurgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Akihiko Teshigawara
- Department of Neurosurgery, The Jikei University School of Medicine Kashiwa Hospital, Chiba, Japan
| | - Takuya Ishii
- Department of Neurosurgery, The Jikei University School of Medicine Daisan Hospital Tokyo, Tokyo, Japan
| | - Yuzuru Hasegawa
- Department of Neurosurgery, The Jikei University School of Medicine Kashiwa Hospital, Chiba, Japan
| | - Yasuharu Akasaki
- Department of Neurosurgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, The Jikei University School of Medicine Kashiwa Hospital, Chiba, Japan; Department of Neurosurgery, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
294
|
Narang A, Gebrael G, Jo Y, Thomas VM, Li H, Fortuna GG, Sayegh N, Tandar C, Tripathi N, Chigarira B, Srivastava A, Hage Chehade C, Nordblad B, Maughan BL, Agarwal N, Swami U. Effectiveness of Second-Line Cabozantinib in Metastatic Clear Cell Renal Cell Carcinoma Patients After First-Line Treatment with Immune Checkpoint Inhibitor-based Combinations. KIDNEY CANCER 2024; 8:135-142. [PMID: 39263256 PMCID: PMC11385087 DOI: 10.3233/kca-240016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/30/2024] [Indexed: 09/13/2024]
Abstract
Background Cabozantinib, a tyrosine kinase inhibitor (TKI), is a prevalent second-line (2 L) therapy and was approved for use after progression on TKIs. However, the 1 L treatment setting has changed since the approval of cabozantinib monotherapy in salvage therapy settings. Objective To assess the differential effectiveness of cabozantinib after prior progression on 1 L ipilimumab with nivolumab (IPI + NIVO) compared to programmed death receptor-1 (PD-1) or PD-1 ligand (PD-L1) inhibitors (PD1/L1i) with TKIs. Methods Utilizing a nationwide electronic health record (EHR)-derived de-identified database, we included patients with metastatic clear cell renal cell carcinoma (mccRCC) who received 1 L treatment with an immune checkpoint inhibitor (ICI)-based combination and 2 L treatment with cabozantinib monotherapy. These patients were categorized based on the type of 1 L ICI-based combination received: IPI + NIVO vs. PD1/L1i with TKI. Real-world time to next therapy (rwTTNT) and real-world overall survival (rwOS) were summarized using Kaplan-Meier curves and compared using Cox-proportional hazard models adjusted for International mRCC Database Consortium (IMDC) risk groups. Results Among 12,285 patients with metastatic renal cell carcinoma, 237 were eligible and included. Median rwTTNT was 8 months for the IPI + NIVO subgroup and 7.5 months for the PD1/L1i + TKI subgroup (HR 1.05, 95% CI: 0.74-1.49, p = 0.8). Median rwOS was 17 months for IPI + NIVO and 16 months for PD1/L1i + TKI subgroup (HR 0.79, 95% CI: 0.52-1.20, p = 0.3). Conclusions Cabozantinib remains effective as a 2 L therapy for mccRCC independent of the type of prior 1 L ICI-based combination. Further research is needed to validate these findings and explore the ideal sequencing of therapies.
Collapse
Affiliation(s)
- Arshit Narang
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Yeonjung Jo
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Vinay Mathew Thomas
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Haoran Li
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, Westwood, KS, USA
| | - Gliceida Galarza Fortuna
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicolas Sayegh
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Clara Tandar
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nishita Tripathi
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Beverly Chigarira
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Ayana Srivastava
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chadi Hage Chehade
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Blake Nordblad
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Benjamin L Maughan
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
295
|
Shapira-Frommer R, Niu J, Perets R, Peters S, Shouse G, Lugowska I, Garassino MC, Sands J, Keenan T, Zhao B, Healy J, Ahn MJ. The KEYVIBE program: vibostolimab and pembrolizumab for the treatment of advanced malignancies. Future Oncol 2024; 20:1983-1991. [PMID: 39230120 PMCID: PMC11497960 DOI: 10.1080/14796694.2024.2343272] [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: 12/12/2023] [Accepted: 04/11/2024] [Indexed: 09/05/2024] Open
Abstract
Vibostolimab, a humanized IgG1 monoclonal antibody, blocks the interaction between TIGIT and its ligands, preventing the immunosuppressive effects of TIGIT. The addition of vibostolimab to the PD-1 inhibitor pembrolizumab has shown promising antitumor activity, warranting further exploration of vibostolimab as a potential therapeutic option. The KEYVIBE program consists of nine trials that will evaluate the safety and efficacy of vibostolimab monotherapy and vibostolimab-based combination therapy in advanced solid tumors and hematological malignancies. These studies will also evaluate coformulated immunotherapy addressing issues that occur with the sequential administration of immunotherapy. The KEYVIBE program will provide further insight into the clinical utility of vibostolimab-based therapy across multiple indications and various stages of disease.
Collapse
Affiliation(s)
- Ronnie Shapira-Frommer
- Division of Oncology and Clinical Research Institute at Rambam, Rambam Medical Center, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, 5265601, Israel
| | - Jiaxin Niu
- Banner MD Anderson Cancer Center, Gilbert, AZ85234, USA
| | - Ruth Perets
- Clinical Research Institute at Rambam, Rambam Medical Center & Technion—Israel Institute of Technology, Haifa, 3109601,Israel
| | - Solange Peters
- Vaud University Hospital Center, University of Lausanne, Lausanne, 1005, Switzerland
| | - Geoffrey Shouse
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA91010, USA
| | - Iwona Lugowska
- Early Phase Clinical Trials Unit, Maria Sklodowska Curie National Research Institute of Oncology, Warsaw, 00-001, Poland
| | | | - Jacob Sands
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA02215, USA
| | | | - Bin Zhao
- Merck & Co., Inc., Rahway, NJ07065, USA
| | | | - Myung-Ju Ahn
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| |
Collapse
|
296
|
Laru L, Ronkainen H, Ohtonen P, Vaarala MH. The impact of metastasectomy on survival of patients with synchronous metastatic renal cell cancer in Finland: A nationwide study. Scand J Surg 2024; 113:219-228. [PMID: 38433655 DOI: 10.1177/14574969241234485] [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] [Indexed: 03/05/2024]
Abstract
BACKGROUND AND OBJECTIVE Most of the studies on metastasectomy in renal cell cancer are based on metachronous, often oligometastatic disease. Prior data on the impact of metastasectomy in synchronous metastatic renal cell cancer (mRCC) is, however, very scarce. We aimed to investigate the role of complete and incomplete metastasectomy in a large, nationwide patient population. METHODS We analyzed nationwide data, including all synchronous mRCC cases in Finland diagnosed during a 6-year period identified from the Finnish Cancer Registry, and complemented with patient records from the treating hospitals. We only included the patients who underwent removal of the primary tumor by nephrectomy. We performed univariate and multivariable adjusted analysis to identify the effect of metastasectomy on overall survival (OS) and cancer-specific survival (CSS). RESULTS We included 483 patients with synchronous mRCC. Overall, 57 patients underwent complete and 96 incomplete metastasectomy, while 330 patients had no metastasectomy. The median OS was 17.9 and CSS 17.2 months for all patients. The median OS and the median CSS were 59.3 and 60.8 months for the complete, 21.9 and 25.1 for the incomplete, and 14.5 and 14.8 months for the no metastasectomy groups (p < 0.001 for differences). In both applied multivariable statistical models, the OS and CSS benefit from complete metastasectomy remained significant (hazard ratios (HRs) varied between 0.42 and 0.54, p < 0.001) compared with the no metastasectomy group. However, there was no improvement in survival estimates in the incomplete metastasectomy group compared with the no metastasectomy group (HRs varied between 1.04 and 1.10, p > 0.40). CONCLUSIONS Complete metastasectomy, when possible, can be considered as a treatment option for selected patients with synchronous mRCC who are fit for surgery. By contrast, we found no survival benefit from an incomplete metastasectomy suggesting that such procedures should not be performed for these patients.
Collapse
Affiliation(s)
- Lauri Laru
- Department of Urology, Oulu University Hospital, P.O. Box 21 FI-90029 OYS, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Hanna Ronkainen
- Department of Urology, Oulu University Hospital, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Pasi Ohtonen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Research Service Unit, Oulu University Hospital, Oulu, Finland
| | - Markku H Vaarala
- Department of Urology, Oulu University Hospital, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
297
|
Campbell MT, Shah AY, Msaouel P, Tannir NM, Siefker-Radtke AO, Kamat AM, Navai N, Dinney CP, Rao P, Guo CC, Sheth RA, Venkatesan AM, Tidwell RS, Yadav SS, Gu A, Chen H, Macaluso M, Duan F, Basu S, Jindal S, Sharma P. A Pilot Study of the CD38 Antagonist Daratumumab in Patients with Metastatic Renal Cell Carcinoma or Muscle-Invasive Bladder Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:2444-2453. [PMID: 39207194 PMCID: PMC11406637 DOI: 10.1158/2767-9764.crc-24-0237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/28/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE We performed a pilot study of daratumumab (an mAb directed against CD38) in muscle-invasive bladder cancer (MIBC) and treatment-refractory metastatic renal cell carcinoma (mRCC). EXPERIMENTAL DESIGN Patients with MIBC underwent baseline transurethral resection of the bladder tumor followed by four weekly doses of daratumumab prior to cystectomy. Patients with mRCC underwent baseline and sequential biopsies after eight weekly doses. The primary endpoint was safety. The secondary endpoints were pathologic complete response rate for the MIBC cohort and objective response rate and progression-free survival for the mRCC cohort. Exploratory analyses included immune monitoring and overall survival. A Bayesian sequential monitoring design for toxicity was used for excessive toxicity. RESULTS In both the MIBC (n = 8) and mRCC (n = 8) cohorts, no toxicity events were encountered. In the MIBC cohort, one patient experienced pathologic complete response rate. In the mRCC cohort, no objective responses were reported, and the median progression-free survival was 1.5 months (95% confidence interval, 1.1-1.8 months). Immune monitoring found significant reductions in NK cells in circulation in both cohorts after treatment. In the tissue analysis, IHC found evidence of diminished CD38 presence in mRCC with treatment, whereas the baseline levels in MIBC were low. CONCLUSION Treatment with daratumumab was safe. No signal of efficacy was detected in mRCC, and conclusions on the activity in MIBC were limited. Evidence of daratumumab targeting CD38 was detected in circulating immune cells and within the tumor microenvironment of mRCC and MIBC. SIGNIFICANCE In this prospective clinical trial of daratumumab, treatment in patients with MIBC and mRCC was safe. Limited efficacy was observed. Treatment with daratumumab resulted in CD38-expressing immune cell subsets to be targeted both in circulation and within the tumor microenvironment.
Collapse
Affiliation(s)
- Matthew T. Campbell
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Amishi Y. Shah
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Nizar M. Tannir
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Arlene O. Siefker-Radtke
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ashish M. Kamat
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Neema Navai
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Colin P.N. Dinney
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Priya Rao
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Charles C. Guo
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Rahul A. Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Aradhana M. Venkatesan
- Division of Diagnostic Imaging, Department of Abdominal Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Rebecca S. Tidwell
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Shalini S. Yadav
- Immunotherapy Platform, University of MD Anderson Cancer Center, Houston, Texas.
| | - Aidi Gu
- Immunotherapy Platform, University of MD Anderson Cancer Center, Houston, Texas.
| | - Hong Chen
- Immunotherapy Platform, University of MD Anderson Cancer Center, Houston, Texas.
- James P. Allison Institute, University of MD Anderson Cancer Center, Houston, Texas.
| | - Marc Macaluso
- Immunotherapy Platform, University of MD Anderson Cancer Center, Houston, Texas.
- James P. Allison Institute, University of MD Anderson Cancer Center, Houston, Texas.
| | - Fei Duan
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Sreyashi Basu
- Immunotherapy Platform, University of MD Anderson Cancer Center, Houston, Texas.
- James P. Allison Institute, University of MD Anderson Cancer Center, Houston, Texas.
| | - Sonali Jindal
- Immunotherapy Platform, University of MD Anderson Cancer Center, Houston, Texas.
- James P. Allison Institute, University of MD Anderson Cancer Center, Houston, Texas.
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
- Immunotherapy Platform, University of MD Anderson Cancer Center, Houston, Texas.
- James P. Allison Institute, University of MD Anderson Cancer Center, Houston, Texas.
- Department of Immunology, University of MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
298
|
Lynch C, Korpics MC, Katipally RR, Bestvina CM, Pitroda SP, Patel JD, Luke JJ, Chmura SJ, Juloori A. Safety of combined ablative radiotherapy and immune checkpoint inhibitors in three phase I trials. Eur J Cancer 2024; 209:114264. [PMID: 39106643 DOI: 10.1016/j.ejca.2024.114264] [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: 05/03/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND Stereotactic body radiotherapy (SBRT) is safe and effective for treatment of extracranial metastatic disease, but its safety when combined with immune checkpoint inhibitors (ICI) has not yet been comprehensively reported. Here we report adverse events (AEs) associated with combined SBRT and ICI using prospectively-collected data on patients in three trials investigating multi-site SBRT combined with ICI. METHODS Patients were included from three prospective trials of ICI (pembrolizumab; nivolumab/urelumab or nivolumab/cabiralizumab; nivolumab/ipilimumab) with SBRT to 1-4 sites. AEs were recorded prospectively using the CTCAE v4.0. Survival was analyzed using Kaplan-Meier method with a 90-day landmark. Association of patient characteristics with cumulative incidence of AEs was assessed using Fine-Gray regression. RESULTS 213 patients were included, with a median follow-up of 10 months. Over the follow-up period, 50 % and 27 % of patients experienced at least one grade ≥ 2 or grade ≥ 3 AE, respectively. Cumulative incidences of grade ≥ 2 and grade ≥ 3 AEs at 6 months were 47 % and 23 %, respectively. Three grade 5 AEs rated "possibly" related to treatment occurred outside the 90-day dose-limiting toxicity window. Landmarked survival analysis of patients with or without grade ≥ 3 AEs showed no significant difference in progression-free or overall survival. Dual-agent ICI was significantly associated with grade ≥ 3 AE. CONCLUSION This analysis features the largest prospectively evaluated cohort of patients treated with combination ablative SBRT and ICI to date and provides context for future trial design. We conclude that multi-site SBRT and ICI can be safely co-administered when SBRT is delivered with prioritization of normal tissue constraints.
Collapse
Affiliation(s)
- Connor Lynch
- The University of Chicago Medical Center, Department of Radiation and Cellular Oncology, United States
| | - Mark C Korpics
- The University of Chicago Medical Center, Department of Radiation and Cellular Oncology, United States
| | - Rohan R Katipally
- The University of Chicago Medical Center, Department of Radiation and Cellular Oncology, United States
| | - Christine M Bestvina
- The University of Chicago Medical Center, Department of Medical Oncology, United States
| | - Sean P Pitroda
- The University of Chicago Medical Center, Department of Radiation and Cellular Oncology, United States
| | - Jyoti D Patel
- Northwestern University, Division of Hematology and Oncology, United States
| | - Jason J Luke
- UPMC Hillman Cancer Center and University of Pittsburgh, United States
| | - Steven J Chmura
- The University of Chicago Medical Center, Department of Radiation and Cellular Oncology, United States
| | - Aditya Juloori
- The University of Chicago Medical Center, Department of Radiation and Cellular Oncology, United States.
| |
Collapse
|
299
|
Brown J, Santini D, Charnley N, Ogareva A, Chisholm A, Jones R. Implications of bone metastasis on response to systemic therapy in patients with advanced renal cell carcinoma: A systematic literature review. Cancer Treat Rev 2024; 129:102792. [PMID: 38972135 DOI: 10.1016/j.ctrv.2024.102792] [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: 02/23/2024] [Revised: 06/07/2024] [Accepted: 06/23/2024] [Indexed: 07/09/2024]
Abstract
INTRODUCTION Bone metastases negatively affect prognosis in patients with advanced renal cell carcinoma (aRCC). We conducted a systematic literature review to identify clinical trial publications including patients with aRCC with and without bone metastases. METHODS The review was conducted according to Preferred Reporting Items for Systematic Reviews and Meta‑Analyses (PRISMA) guidelines and registered with PROSPERO (CRD42022355436). MEDLINE and Embase databases were searched (September 2, 2022) to identify publications reporting efficacy and safety outcomes for patients with/without bone metastasis from clinical trials of systemic RCC therapies. Risk of bias was assessed using Grading of Recommendations Assessment, Development and Evaluation (GRADE). RESULTS Of 526 publications screened, 19 were eligible: seven (from five studies) reported phase 3 trials, six reported phase 2 trials, one reported phase 1b/2 trials, and five were pooled analyses. Five publications reported moderate-quality evidence, while 14 were graded as low- or very low-quality evidence, suggesting a high potential for uncertainty. Five studies reported benefits of investigational therapies versus comparators in patients with and without bone metastases; these studies included cabozantinib, nivolumab, cabozantinib plus nivolumab, and lenvatinib plus pembrolizumab treatment arms. Data were also available for nivolumab plus ipilimumab. Bone metastases were consistently associated with poor prognosis in patients with aRCC. Preliminary data support the hypothesis that therapies targeting pathways implicated in the development of bone metastases may be beneficial, and warrant further investigation. However, data to support treatment decision-making are lacking. CONCLUSION Our findings highlight the need for clinical data to assist in defining the optimal treatment for patients with aRCC and bone metastasis.
Collapse
Affiliation(s)
- Janet Brown
- Division of Clinical Medicine, University of Sheffield, Sheffield, UK.
| | - Daniele Santini
- Medical Oncology A, Policlinico Umberto 1, La Sapienza University of Rome, Italy
| | | | | | | | - Robert Jones
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
300
|
Cordial P, Bentley ID, Horowitz JC, Ho K. Eosinophilic reactive airways disease after immune checkpoint inhibitor treatment. Respirol Case Rep 2024; 12:e70022. [PMID: 39253321 PMCID: PMC11381182 DOI: 10.1002/rcr2.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) are increasingly utilized as first-line treatment for many solid tumour malignancies. One downside of ICI therapy is autoimmune-mediated organ inflammation, or immune-related adverse events (irAE). ICI-related pneumonitis, or non-infectious inflammation of the lung, is a well-described irAE. While guidelines surrounding ICI-related pneumonitis are well established, other ICI-related pulmonary toxicities, including reactive airways disease, are rarely described in the literature. Here, we present a series of patients without pre-existing COPD or asthma who developed reactive airways disease with peripheral eosinophilia after ICI therapy and without radiographic evidence of pneumonitis. The patients were treated with typical therapies for reactive airways disease, including- inhaled steroids, bronchodilators, systemic steroids, and in one instance, dupilumab. All experienced symptomatic improvement with these therapies, enabling some of the patients to continue receiving ICI therapy.
Collapse
Affiliation(s)
- Parker Cordial
- Internal Medicine The Ohio State University Wexner Medical Center Columbus Ohio USA
| | - Ian D Bentley
- Division of Pulmonary, Critical Care and Sleep Medicine The Ohio State University Wexner Medical Center Columbus Ohio USA
- Dorothy M. Davis Heart and Lung Research Institute The Ohio State Wexner Medical Center Columbus Ohio USA
| | - Jeffrey C Horowitz
- Division of Pulmonary, Critical Care and Sleep Medicine The Ohio State University Wexner Medical Center Columbus Ohio USA
- Dorothy M. Davis Heart and Lung Research Institute The Ohio State Wexner Medical Center Columbus Ohio USA
| | - Kevin Ho
- Division of Pulmonary, Critical Care and Sleep Medicine The Ohio State University Wexner Medical Center Columbus Ohio USA
- Dorothy M. Davis Heart and Lung Research Institute The Ohio State Wexner Medical Center Columbus Ohio USA
| |
Collapse
|