1
|
Akbar S, Setia T, Das S, Kumari S, Rahaman SB, Wasim M, Ahmed B, Dewangan RP. Design, synthesis, and evaluation of 1,4-benzodioxane-hydrazone derivatives as potential therapeutics for skin cancer: In silico, in vitro, and in vivo studies. Bioorg Chem 2025; 160:108449. [PMID: 40220711 DOI: 10.1016/j.bioorg.2025.108449] [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: 01/01/2025] [Revised: 03/26/2025] [Accepted: 04/06/2025] [Indexed: 04/14/2025]
Abstract
In the pursuit of novel chemotherapeutic agents for skin cancer, we synthesized a series of 1,4-benzodioxane-hydrazone derivatives (7a-l) using the Wolff-Kishner reaction. These compounds were initially screened against the NCI-60 oncological cell lines in a one-dose assay at 10 μM. Among them, compound 7e emerged as a potent inhibitor of cancer cell growth across 56 cell lines, with an average GI50 of 6.92 μM. Notably, it exhibited enhanced efficacy in melanoma cell lines, including MDA-MB-435, M14, SK-MEL-2, and UACC-62, with GI50 values of 0.20, 0.46, 0.57, and 0.27 μM, respectively. Apoptosis assay and cell cycle analysis studies revealed that compound 7e induced apoptosis and caused S-phase arrest in MDA-MB-435 cells. Furthermore, an in vitro enzyme inhibition assay against mTOR kinase yielded an IC50 of 5.47 μM, while molecular docking studies of compound 7e (docking score: -8.105 kcal/mol) supported its binding affinity. Compound 7e adhered to Lipinski's rule of five and displayed favourable ADMET properties. In vivo studies demonstrated its safety and efficacy in ameliorating skin cancer in a mice model when administered intraperitoneally at 20 mg/kg. Structure-activity relationships were established through in vitro, in vivo, molecular docking, and molecular dynamics analysis. Collectively, these findings highlight 1,4-benzodioxane-hydrazone derivatives as promising scaffold for the development of novel chemotherapeutic agents for skin cancer.
Collapse
Affiliation(s)
- Saleem Akbar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed to be University), New Delhi 110062, India
| | - Tushar Setia
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed to be University), New Delhi 110062, India
| | - Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shalini Kumari
- CSIR-Institute of Genomics and Integrative Biology (IGIB), Sukhdev Vihar, Mathura Road, New Delhi 110025, India
| | - Sk Batin Rahaman
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed to be University), New Delhi 110062, India
| | - Mohd Wasim
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed to be University), New Delhi 110062, India
| | - Bahar Ahmed
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed to be University), New Delhi 110062, India.
| | - Rikeshwer Prasad Dewangan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed to be University), New Delhi 110062, India.
| |
Collapse
|
2
|
Braun DA, Moranzoni G, Chea V, McGregor BA, Blass E, Tu CR, Vanasse AP, Forman C, Forman J, Afeyan AB, Schindler NR, Liu Y, Li S, Southard J, Chang SL, Hirsch MS, LeBoeuf NR, Olive O, Mehndiratta A, Greenslade H, Shetty K, Klaeger S, Sarkizova S, Pedersen CB, Mossanen M, Carulli I, Tarren A, Duke-Cohan J, Howard AA, Iorgulescu JB, Shim B, Simon JM, Signoretti S, Aster JC, Elagina L, Carr SA, Leshchiner I, Getz G, Gabriel S, Hacohen N, Olsen LR, Oliveira G, Neuberg DS, Livak KJ, Shukla SA, Fritsch EF, Wu CJ, Keskin DB, Ott PA, Choueiri TK. A neoantigen vaccine generates antitumour immunity in renal cell carcinoma. Nature 2025; 639:474-482. [PMID: 39910301 PMCID: PMC11903305 DOI: 10.1038/s41586-024-08507-5] [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: 02/19/2024] [Accepted: 12/10/2024] [Indexed: 02/07/2025]
Abstract
Personalized cancer vaccines (PCVs) can generate circulating immune responses against predicted neoantigens1-6. However, whether such responses can target cancer driver mutations, lead to immune recognition of a patient's tumour and result in clinical activity are largely unknown. These questions are of particular interest for patients who have tumours with a low mutational burden. Here we conducted a phase I trial (ClinicalTrials.gov identifier NCT02950766) to test a neoantigen-targeting PCV in patients with high-risk, fully resected clear cell renal cell carcinoma (RCC; stage III or IV) with or without ipilimumab administered adjacent to the vaccine. At a median follow-up of 40.2 months after surgery, none of the 9 participants enrolled in the study had a recurrence of RCC. No dose-limiting toxicities were observed. All patients generated T cell immune responses against the PCV antigens, including to RCC driver mutations in VHL, PBRM1, BAP1, KDM5C and PIK3CA. Following vaccination, there was a durable expansion of peripheral T cell clones. Moreover, T cell reactivity against autologous tumours was detected in seven out of nine patients. Our results demonstrate that neoantigen-targeting PCVs in high-risk RCC are highly immunogenic, capable of targeting key driver mutations and can induce antitumour immunity. These observations, in conjunction with the absence of recurrence in all nine vaccinated patients, highlights the promise of PCVs as effective adjuvant therapy in RCC.
Collapse
Affiliation(s)
- David A Braun
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Giorgia Moranzoni
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Vipheaviny Chea
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Bradley A McGregor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Eryn Blass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Chloe R Tu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Allison P Vanasse
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Cleo Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Juliet Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexander B Afeyan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nicholas R Schindler
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Yiwen Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuqiang Li
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jackson Southard
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven L Chang
- Harvard Medical School, Boston, MA, USA
- Department of Urology, Brigham and Women's Hospital, Boston, MA, USA
| | - Michelle S Hirsch
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Nicole R LeBoeuf
- Harvard Medical School, Boston, MA, USA
- Center for Cutaneous Oncology, Dana-Farber Brigham and Women's Cancer Center, Boston, MA, USA
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, USA
| | - Oriol Olive
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ambica Mehndiratta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Haley Greenslade
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Keerthi Shetty
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susan Klaeger
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Christina B Pedersen
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Center for Genomic Medicine, Rigshospitalet-Copenhagen University Hospital, Copenhagen, Denmark
| | - Matthew Mossanen
- Harvard Medical School, Boston, MA, USA
- Department of Urology, Brigham and Women's Hospital, Boston, MA, USA
| | - Isabel Carulli
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anna Tarren
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joseph Duke-Cohan
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexis A Howard
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - J Bryan Iorgulescu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bohoon Shim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jeremy M Simon
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jon C Aster
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ignaty Leshchiner
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, USA
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Lars R Olsen
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Donna S Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Livak
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sachet A Shukla
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edward F Fritsch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Bedke J, Grünwald V. [Adjuvant therapy for renal cell carcinoma : Relevant patient and tumor factors]. UROLOGIE (HEIDELBERG, GERMANY) 2024:10.1007/s00120-024-02474-5. [PMID: 39545946 DOI: 10.1007/s00120-024-02474-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 11/17/2024]
Abstract
The gold standard in the treatment of localized and locally advanced renal cell carcinoma is surgery. Nevertheless, there is still a risk of tumor relapse. Reducing the risk of recurrence and extending overall survival is the goal of subsequent adjuvant treatment. The aim of this work is to discuss the current and future landscape of adjuvant therapy, taking into account the risk-benefit balance in the individual patient selected for adjuvant treatment. The immune checkpoint inhibitor (CPI) pembrolizumab demonstrated a significant increase in disease-free and overall survival after surgery for the first time. However, other CPI studies demonstrated no improvement. Patient selection for adjuvant treatment is currently based on the parameters of the TNM system. Prospective biomarkers are currently not available. Here, kidney injury molecule‑1 (KIM-1) represents an initial promising biomarker in the prediction of adjuvant immunotherapy.
Collapse
Affiliation(s)
- Jens Bedke
- Klinik für Urologie & Transplantationschirurgie, Eva Mayr-Stihl Cancer Center Stuttgart, Klinikum Stuttgart, Kriegsbergstraße 60, 70191, Stuttgart, Deutschland.
| | - Viktor Grünwald
- Klinik für Innere Medizin (Tumorforschung) und Klinik für Urologie, Westdeutsches Tumorzentrum Essen, Universitätsklinikum Essen, Essen, Deutschland
| |
Collapse
|
4
|
Kasherman L, Siu DHW, Woodford R, Harris CA. Angiogenesis Inhibitors and Immunomodulation in Renal Cell Cancers: The Past, Present, and Future. Cancers (Basel) 2022; 14:1406. [PMID: 35326557 PMCID: PMC8946206 DOI: 10.3390/cancers14061406] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis inhibitors have been adopted into the standard armamentarium of therapies for advanced-stage renal cell carcinomas (RCC), but more recently, combination regimens with immune checkpoint inhibitors have demonstrated better outcomes. Despite this, the majority of affected patients still eventually experience progressive disease due to therapeutic resistance mechanisms, and there remains a need to develop novel therapeutic strategies. This article will review the synergistic mechanisms behind angiogenesis and immunomodulation in the tumor microenvironment and discuss the pre-clinical and clinical evidence for both clear-cell and non-clear-cell RCC, exploring opportunities for future growth in this exciting area of drug development.
Collapse
Affiliation(s)
- Lawrence Kasherman
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2217, Australia
- Department of Medical Oncology, Illawarra Cancer Care Centre, Wollongong, NSW 2500, Australia
| | - Derrick Ho Wai Siu
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- National Health Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW 2050, Australia
| | - Rachel Woodford
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- Faculty of Medciine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Carole A. Harris
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2217, Australia
| |
Collapse
|
5
|
Amores Bermúdez J, Osman García I, Unda Urzáiz M, Jiménez Marrero P, Ledo Cepero MJ, Llarena R, Flores Martín J, Abad Vivas-Pérez JI, Rodrigo Aliaga M, Juarez Soto A. Safety of nivolumab in metastatic renal cell carcinoma patients: A real-life experience in a Spanish urology setting. Actas Urol Esp 2019; 43:364-370. [PMID: 31097211 DOI: 10.1016/j.acuro.2019.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/12/2019] [Accepted: 03/04/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION AND OBJECTIVES Nivolumab is an immunotherapy agent that has been an approved treatment for previously treated patients with advanced renal cell carcinoma (RCC). Experience in real-life settings, especially regarding immune- related adverse events, is scarce. We present our experience with reference to the safety of nivolumab in patients with metastatic RCC (mRCC) treated in 9 hospitals in Spain. MATERIAL AND METHODS Retrospective, multicentre study of patients with mRCC treated with nivolumab between 2016 and 2018. Data on baseline socio-demographic and clinical characteristics and drug-related adverse events were collected. RESULTS The mean age of the 26 patients included was 63.7±11.5 years; 96% were ECOG 0-1 and 78% had favourable or intermediate MSKCC risk scores; 73% had the clear cell histological subtype and 30% metastatic disease. Median follow-up was 9 months (range 1-14). All patients experienced an adverse event at different grades, with fatigue, fever and anaemia being the most common (27%). Grade 3 adverse events occurred in 23% of patients. Adverse reactions led to treatment suspension in 3 patients (11%). CONCLUSION In the real-life clinical setting, nivolumab shows favourable outcomes, similar to those reported by other studies.
Collapse
Affiliation(s)
- J Amores Bermúdez
- Servicio de Urología, Hospital Universitario de Jerez, Jerez, España.
| | - I Osman García
- Servicio de Urología, Hospital Virgen del Rocío, Sevilla, España
| | - M Unda Urzáiz
- Servicio de Urología, Hospital Universitario Basurto, Bilbao, España
| | - P Jiménez Marrero
- Servicio de Urología, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria, España
| | - M J Ledo Cepero
- Servicio de Urología, Hospital Universitario Puerta del Mar, Cádiz, España
| | - R Llarena
- Servicio de Urología, Hospital de Cruces, Baracaldo, España
| | - J Flores Martín
- Servicio de Urología, Complejo Hospitalario de Jaén, Jaén, España
| | | | - M Rodrigo Aliaga
- Servicio de Urología, Hospital General Universitario de Castellón, Castellón, España
| | - A Juarez Soto
- Servicio de Urología, Hospital Universitario de Jerez, Jerez, España
| |
Collapse
|
6
|
Limitations to the Therapeutic Potential of Tyrosine Kinase Inhibitors and Alternative Therapies for Kidney Cancer. Ochsner J 2019; 19:138-151. [PMID: 31258426 DOI: 10.31486/toj.18.0015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Renal cell carcinomas (RCCs) are the most common primary renal tumor. RCCs have a high rate of metastasis and have the highest mortality rate of all genitourinary cancers. They are often diagnosed late when metastases have developed, and these metastases are difficult to treat successfully. Since 2006, the standard first-line treatment for patients with metastatic RCC has been multitargeted tyrosine kinase inhibitors (TKIs) that include mammalian target of rapamycin (mTOR) inhibitors. RCCs are highly vascularized tumors, and their angiogenesis is controlled by tyrosine kinases that play a vital role in growth factor signaling to stimulate this process. TKI therapy was introduced for direct targeting of angiogenesis in RCC. TKIs have been moderately successful in the treatment of metastatic RCC and initially increased cancer-specific survival times. However, RCC rapidly becomes resistant to TKIs, and no current drug has produced a cure for advanced RCC. Methods: We provide an overview of RCC, explain some reasons for therapy resistance in RCC, and describe some therapies that may overcome resistance to TKIs. The key pathways that determine therapy resistance are illustrated. Results: Factors involved in the development and progression of RCC include genetic mutations, activation of hypoxia-inducible factor and related proteins, cellular metabolism, the tumor microenvironment, and growth factors and their receptors. Resistance to the therapeutic potential of TKIs can be acquired or intrinsic. Alternative therapies include other small molecule drugs and immunotherapy based on immune checkpoint blockade. Conclusion: The treatment of RCC is undergoing a paradigm shift from sole use of small molecule antiangiogenesis TKIs as first-line therapy to include newly approved agents for second-line and third-line therapy that now involve the mTOR pathway and immune checkpoint blockade drugs for patients with advanced RCC.
Collapse
|
7
|
Circulating Tumor Cells for the Management of Renal Cell Carcinoma. Diagnostics (Basel) 2018; 8:diagnostics8030063. [PMID: 30177639 PMCID: PMC6164661 DOI: 10.3390/diagnostics8030063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022] Open
Abstract
Renal cell carcinoma is a highly malignant cancer that would benefit from non-invasive innovative markers providing early diagnosis and recurrence detection. Circulating tumor cells are a particularly promising marker of tumor invasion that could be used to improve the management of patients with RCC. However, the extensive genetic and immunophenotypic heterogeneity of cells from RCC and their trend to transition to the mesenchymal phenotype when they circulate in blood constitute a challenge for their sensitive and specific detection. This review analyzes published studies targeting CTC in patients with RCC, in the context of the biological, pathological, and molecular complexity of this particular cancer. Although further analytical and clinical studies are needed to pinpoint the most suitable approach for highly sensitive CTC detection in RCC patients, it is clear that this field can bring a relevant guide to clinicians and help to RCC patients. Furthermore, as described, a particular subtype of RCC-the ccRCC-can be used as a model to study the relationship between cytomorphological and genetic cellular markers of malignancy, an important issue for the study of CTC from any type of solid cancer.
Collapse
|
8
|
Zhang XY, Elfarra AA. Toxicity mechanism-based prodrugs: glutathione-dependent bioactivation as a strategy for anticancer prodrug design. Expert Opin Drug Discov 2018; 13:815-824. [PMID: 30101640 DOI: 10.1080/17460441.2018.1508207] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION 6-Mercaptopurine (6-MP) and 6-thioguanine (6-TG), two anticancer drugs, have high systemic toxicity due to a lack of target specificity. Therefore, increasing target selectivity should improve drug safety. Areas covered: The authors examined the hypothesis that new prodrug designs based upon mechanisms of kidney-selective toxicity of trichloroethylene would reduce systemic toxicity and improve selectivity to kidney and tumor cells. Two approaches specifically were investigated. The first approach was based upon bioactivation of trichloroethylene-cysteine S-conjugate by renal cysteine S-conjugate β-lyases. The prodrugs obtained were kidney-selective but exhibited low turnover rates. The second approach was based on the toxic mechanism of trichloroethylene-cysteine S-conjugate sulfoxide, a Michael acceptor that undergoes rapid addition-elimination reactions with biological thiols. Expert opinion: Glutathione-dependent Michael addition-elimination reactions appear to be an excellent strategy to design highly efficient anticancer drugs. Targeting glutathione could be a promising approach for the development of anticancer prodrugs because cancer cells usually upregulate glutathione biosynthesis and/or glutathione S-transferases expression.
Collapse
Affiliation(s)
- Xin-Yu Zhang
- a Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Faculty of Public Health , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Adnan A Elfarra
- b Department of Comparative Biosciences and the Molecular and Environmental Toxicology Center , University of Wisconsin-Madison , Madison , WI , USA
| |
Collapse
|
9
|
|
10
|
Results of a Phase 1/2 Study in Metastatic Renal Cell Carcinoma Patients Treated with a Patient-specific Adjuvant Multi-peptide Vaccine after Resection of Metastases. Eur Urol Focus 2017; 5:604-607. [PMID: 28988765 DOI: 10.1016/j.euf.2017.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/13/2017] [Indexed: 11/21/2022]
Abstract
Treatment of metastatic renal cell carcinoma comprises metastasectomy±systemic medical treatment. Specific immunotherapy after metastasectomy could be a complementary option. In this phase 1/2 study, safety and tolerability of an adjuvant multi-peptide vaccine (UroRCC) after metastasectomy was evaluated together with immune response and efficacy, compared with a contemporary cohort of patients (n=44) treated with metastasectomy only. Nineteen metastatic renal cell carcinoma patients received UroRCC via intradermal or subcutaneous application randomized to immunoadjuvants (granulocyte-macrophage colony-stimulating factor or Montanide). Adverse events of UroRCC were mainly grade I and II; frequency of immune response was higher for major histocompatibility complex class II peptides (17/19, 89.5%) than for major histocompatibility complex class I peptides (8/19, 42.1%). Median overall survival was not reached in the UroRCC group (mean: 112.6 mo, 95% confidence interval [CI]: 92.1-133.1) and 58.0 mo (95% CI: 32.7-83.2) in the control cohort (p=0.015). UroRCC was an independent prognosticator of overall survival (hazard ratio=0.19, 95% CI: 0.05-0.69, p=0.012). Adjuvant UroRCC multi-peptide vaccine after metastasectomy was well tolerated, immunogenic, and indicates potential clinical benefit when compared with a contemporary control cohort (NCT02429440). PATIENT SUMMARY: The application of a patient-specific peptide vaccine after complete resection of metastases in metastatic renal cell carcinoma patients resulted in favorable tolerability and outcome.
Collapse
|
11
|
Bedke J, Stenzl A, Rausch S. AGS-003 combined with sunitinib for the precision treatment of metastatic renal cell carcinoma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1375852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Jens Bedke
- Department of Urology, Eberhard Karls University, Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, Eberhard Karls University, Tübingen, Germany
| | - Steffen Rausch
- Department of Urology, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
12
|
Unverzagt S, Moldenhauer I, Nothacker M, Roßmeißl D, Hadjinicolaou AV, Peinemann F, Greco F, Seliger B. Immunotherapy for metastatic renal cell carcinoma. Cochrane Database Syst Rev 2017; 5:CD011673. [PMID: 28504837 PMCID: PMC6484451 DOI: 10.1002/14651858.cd011673.pub2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Since the mid-2000s, the field of metastatic renal cell carcinoma (mRCC) has experienced a paradigm shift from non-specific therapy with broad-acting cytokines to specific regimens, which directly target the cancer, the tumour microenvironment, or both.Current guidelines recommend targeted therapies with agents such as sunitinib, pazopanib or temsirolimus (for people with poor prognosis) as the standard of care for first-line treatment of people with mRCC and mention non-specific cytokines as an alternative option for selected patients.In November 2015, nivolumab, a checkpoint inhibitor directed against programmed death-1 (PD-1), was approved as the first specific immunotherapeutic agent as second-line therapy in previously treated mRCC patients. OBJECTIVES To assess the effects of immunotherapies either alone or in combination with standard targeted therapies for the treatment of metastatic renal cell carcinoma and their efficacy to maximize patient benefit. SEARCH METHODS We searched the Cochrane Library, MEDLINE (Ovid), Embase (Ovid), ISI Web of Science and registers of ongoing clinical trials in November 2016 without language restrictions. We scanned reference lists and contacted experts in the field to obtain further information. SELECTION CRITERIA We included randomized controlled trials (RCTs) and quasi-RCTs with or without blinding involving people with mRCC. DATA COLLECTION AND ANALYSIS We collected and analyzed studies according to the published protocol. Summary statistics for the primary endpoints were risk ratios (RRs) and mean differences (MD) with their 95% confidence intervals (CIs). We rated the quality of evidence using GRADE methodology and summarized the quality and magnitude of relative and absolute effects for each primary outcome in our 'Summary of findings' tables. MAIN RESULTS We identified eight studies with 4732 eligible participants and an additional 13 ongoing studies. We categorized studies into comparisons, all against standard therapy accordingly as first-line (five comparisons) or second-line therapy (one comparison) for mRCC.Interferon (IFN)-α monotherapy probably increases one-year overall mortality compared to standard targeted therapies with temsirolimus or sunitinib (RR 1.30, 95% CI 1.13 to 1.51; 2 studies; 1166 participants; moderate-quality evidence), may lead to similar quality of life (QoL) (e.g. MD -5.58 points, 95% CI -7.25 to -3.91 for Functional Assessment of Cancer - General (FACT-G); 1 study; 730 participants; low-quality evidence) and may slightly increase the incidence of adverse events (AEs) grade 3 or greater (RR 1.17, 95% CI 1.03 to 1.32; 1 study; 408 participants; low-quality evidence).There is probably no difference between IFN-α plus temsirolimus and temsirolimus alone for one-year overall mortality (RR 1.13, 95% CI 0.95 to 1.34; 1 study; 419 participants; moderate-quality evidence), but the incidence of AEs of 3 or greater may be increased (RR 1.30, 95% CI 1.17 to 1.45; 1 study; 416 participants; low-quality evidence). There was no information on QoL.IFN-α alone may slightly increase one-year overall mortality compared to IFN-α plus bevacizumab (RR 1.17, 95% CI 1.00 to 1.36; 2 studies; 1381 participants; low-quality evidence). This effect is probably accompanied by a lower incidence of AEs of grade 3 or greater (RR 0.77, 95% CI 0.71 to 0.84; 2 studies; 1350 participants; moderate-quality evidence). QoL could not be evaluated due to insufficient data.Treatment with IFN-α plus bevacizumab or standard targeted therapy (sunitinib) may lead to similar one-year overall mortality (RR 0.37, 95% CI 0.13 to 1.08; 1 study; 83 participants; low-quality evidence) and AEs of grade 3 or greater (RR 1.18, 95% CI 0.85 to 1.62; 1 study; 82 participants; low-quality evidence). QoL could not be evaluated due to insufficient data.Treatment with vaccines (e.g. MVA-5T4 or IMA901) or standard therapy may lead to similar one-year overall mortality (RR 1.10, 95% CI 0.91 to 1.32; low-quality evidence) and AEs of grade 3 or greater (RR 1.16, 95% CI 0.97 to 1.39; 2 studies; 1065 participants; low-quality evidence). QoL could not be evaluated due to insufficient data.In previously treated patients, targeted immunotherapy (nivolumab) probably reduces one-year overall mortality compared to standard targeted therapy with everolimus (RR 0.70, 95% CI 0.56 to 0.87; 1 study; 821 participants; moderate-quality evidence), probably improves QoL (e.g. RR 1.51, 95% CI 1.28 to 1.78 for clinically relevant improvement of the FACT-Kidney Symptom Index Disease Related Symptoms (FKSI-DRS); 1 study, 704 participants; moderate-quality evidence) and probably reduces the incidence of AEs grade 3 or greater (RR 0.51, 95% CI 0.40 to 0.65; 1 study; 803 participants; moderate-quality evidence). AUTHORS' CONCLUSIONS Evidence of moderate quality demonstrates that IFN-α monotherapy increases mortality compared to standard targeted therapies alone, whereas there is no difference if IFN is combined with standard targeted therapies. Evidence of low quality demonstrates that QoL is worse with IFN alone and that severe AEs are increased with IFN alone or in combination. There is low-quality evidence that IFN-α alone increases mortality but moderate-quality evidence on decreased AEs compared to IFN-α plus bevacizumab. Low-quality evidence shows no difference for IFN-α plus bevacizumab compared to sunitinib with respect to mortality and severe AEs. Low-quality evidence demonstrates no difference of vaccine treatment compared to standard targeted therapies in mortality and AEs, whereas there is moderate-quality evidence that targeted immunotherapies reduce mortality and AEs and improve QoL.
Collapse
Affiliation(s)
- Susanne Unverzagt
- Martin Luther University Halle‐WittenbergInstitute of Medical Epidemiology, Biostatistics and InformaticsMagdeburge Straße 8Halle/SaaleGermany06097
| | - Ines Moldenhauer
- Martin Luther University Halle‐WittenbergGartenstadtstrasse 22Halle/SaaleGermany06126
| | | | - Dorothea Roßmeißl
- Martin Luther University Halle‐WittenbergMedical FacultyHoher Weg 6Halle/SaaleGermany06120
| | - Andreas V Hadjinicolaou
- University of OxfordHuman Immunology Unit, Institute of Molecular Medicine, Radcliffe Department of
MedicineMerton College, Merton StreetOxfordUKOX1 4JD
| | - Frank Peinemann
- Children's Hospital, University of ColognePediatric Oncology and HematologyKerpener Str. 62CologneGermany50937
| | - Francesco Greco
- Martin Luther University Halle‐WittenbergDepartment of Urology and Renal TransplantationErnst‐Grube‐Strasse 40Halle/SaaleGermany06120
| | - Barbara Seliger
- Martin Luther University Halle‐WittenbergInstitute of Medical ImmunologyHalle/SaaleGermany
| |
Collapse
|
13
|
Bedke J, Gauler T, Grünwald V, Hegele A, Herrmann E, Hinz S, Janssen J, Schmitz S, Schostak M, Tesch H, Zastrow S, Miller K. Systemic therapy in metastatic renal cell carcinoma. World J Urol 2017; 35:179-188. [PMID: 27277600 PMCID: PMC5272893 DOI: 10.1007/s00345-016-1868-5] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/24/2016] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Current systemic treatment of targeted therapies, namely the vascular endothelial growth factor-antibody (VEGF-AB), VEGF receptor tyrosine kinase inhibitor (TKI) and mammalian target of rapamycin (mTOR) inhibitors, have improved progression-free survival and replaced non-specific immunotherapy with cytokines in metastatic renal cell carcinoma (mRCC). METHODS A panel of experts convened to review currently available phase 3 data for mRCC treatment of approved agents, in addition to available EAU guideline data for a collaborative review as the plurality of substances offers different options of first-, second- and third-line treatment with potential sequencing. RESULTS Sunitinib and pazopanib are approved treatments in first-line therapy for patients with favorable- or intermediate-risk clear cell RCC (ccRCC). Temsirolimus has proven benefit over interferon-alfa (IFN-α) in patients with non-clear cell RCC (non-ccRCC). In the second-line treatment TKIs or mTOR inhibitors are treatment choices. Therapy options after TKI failure consist of everolimus and axitinib. Available third-line options consist of everolimus and sorafenib. Recently, nivolumab, a programmed death-1 (PD1) checkpoint inhibitor, improved overall survival benefit compared to everolimus after failure of one or two VEGFR-targeted therapies, which is likely to become the first established checkpoint inhibitor in mRCC. Data for the sequencing of agents remain limited. CONCLUSIONS Despite the high level of evidence for first and second-line treatment in mRCC, data for third-line therapy are limited. Possible sequences include TKI-mTOR-TKI or TKI-TKI-mTOR with the upcoming checkpoint inhibitors in perspective, which might settle a new standard of care after previous TKI therapy.
Collapse
Affiliation(s)
- Jens Bedke
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Strasse 3, 72076, Tübingen, Germany.
| | - Thomas Gauler
- Department of Radiation Oncology, University of Essen, Essen, Germany
| | - Viktor Grünwald
- Department of Hematology and Oncology, Medical School Hannover, Hannover, Germany
| | - Axel Hegele
- Department of Urology and Pediatric Urology, University of Marburg, Marburg, Germany
| | - Edwin Herrmann
- Department of Urology, University of Münster, Münster, Germany
| | - Stefan Hinz
- Department of Urology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | | | - Stephan Schmitz
- Gemeinschaftspraxis für Onkologie und Hämatologie, Köln, Germany
| | - Martin Schostak
- Department of Urology, University of Magdeburg, Magdeburg, Germany
| | - Hans Tesch
- Onkologie Bethanien, Frankfurt am Main, Germany
| | - Stefan Zastrow
- Department of Urology, Technical University of Dresden, Dresden, Germany
| | - Kurt Miller
- Department of Urology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| |
Collapse
|
14
|
Hirbod-Mobarakeh A, Gordan HA, Zahiri Z, Mirshahvalad M, Hosseinverdi S, Rini BI, Rezaei N. Specific immunotherapy in renal cancer: a systematic review. Ther Adv Urol 2016; 9:45-58. [PMID: 28203287 DOI: 10.1177/1756287216681246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Renal cell cancer (RCC) is the tenth most common malignancy in adults. In recent years, several approaches of active and passive immunotherapy have been studied extensively in clinical trials of patients with RCC. The aim of this systematic review was to assess the clinical efficacy of various approaches of specific immunotherapy in patients with RCC. METHODS We searched Medline, Scopus, CENTRAL, TRIP, DART, OpenGrey and ProQuest without any language filter through to 9 October 2015. One author reviewed search results for irrelevant and duplicate studies and two other authors independently extracted data from the studies. We collated study findings and calculated a weighted treatment effect across studies using Review Manager (version 5.3. Copenhagen: The Nordic Cochrane Centre, the Cochrane Collaboration). RESULTS We identified 14 controlled studies with 4013 RCC patients after excluding irrelevant and duplicate studies from 11,319 references retrieved from a literature search. Overall, five autologous tumor cell vaccines, one peptide-based vaccine, one virus-based vaccine and one dendritic cell (DC)-based vaccine were studied in nine controlled studies of active specific immunotherapies. A total of three passive immunotherapies including autologous cytokine-induced killer (CIK) cells, auto lymphocyte therapy (ALT) and autologous lymphokine-activated killer (LAK) cells were studied in four controlled studies. The clinical efficacy of tumor lysate-pulsed DCs, with CIK cells was studied in one controlled trial concurrently. The overall quality of studies was fair. Meta-analysis of seven studies showed that patients undergoing specific immunotherapy had significantly higher overall survival (OS) than those in the control group [hazard ratio (HR) = 0.72; 95% confidence interval (CI) = 0.58-0.89, p = 0.003]. In addition, a meta-analysis of four studies showed that there was a significant difference in progression-free survival (PFS) between patients undergoing specific immunotherapy and patients in control groups (HR = 0.86; 95% CI = 0.73-1, p = 0.05). CONCLUSIONS Results of this systematic review suggest that some specific immunotherapies such as Reniale, ACHN-IL-2, Newcastle disease virus (NDV) virus-infected autologous tumor cells, ALT and CIK treatment could be beneficiary for the treatment of patients with RCC.
Collapse
Affiliation(s)
- Armin Hirbod-Mobarakeh
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran Molecular Immunology Research Center and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hesam Addin Gordan
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zahra Zahiri
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Mirshahvalad
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sima Hosseinverdi
- Border of Immune Tolerance Education and Research Network (BITERN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Brian I Rini
- Department of Solid Tumor Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Dr Qarib Street, Keshavarz Boulevard, Tehran 14194, Iran
| |
Collapse
|
15
|
Makboul R, Refaiy A, Abdelkawi IF, Hameed D, Elderwy AA, Shalaby MM, Merseburger AS, Hussein MRA. Alterations of mTOR and PTEN protein expression in schistosomal squamous cell carcinoma and urothelial carcinoma. Pathol Res Pract 2016; 212:385-92. [DOI: 10.1016/j.prp.2016.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/02/2015] [Accepted: 02/01/2016] [Indexed: 12/23/2022]
|
16
|
B7-H1/PD-1 blockade therapy in urological malignancies: current status and future prospects. TUMORI JOURNAL 2015; 101:549-54. [PMID: 26045125 DOI: 10.5301/tj.5000326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2015] [Indexed: 01/06/2023]
Abstract
The stimulatory and inhibitory coreceptors expressed by T lymphocytes are known to play critical roles in regulating cancer immunity. An array of inhibitory coreceptors involved in the inhibition of T-cell functions and the blockade of immune activation have been discovered in recent years, the most important of which are cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmmed death-1 (PD-1), and B7 homolog 1 (B7-H1). Immunotherapies targeting T-cell coinhibitory molecules have proved to be effective in cancer treatment. Several kinds of monoclonal antibodies have been tested in preclinical studies, with better outcomes than conventional therapies in many malignancies. Common urological malignancies including renal cell carcinoma, bladder cancer and prostate cancer are supposed to be immunogenic cancer types and not so sensitive to conventional therapies as other malignancies. This review will focus on B7-H1/PD-1 blockade therapy in urological malignancies, summarizing the results of clinical trials as well as the challenges and prospects of this emerging immunotherapy.
Collapse
|
17
|
Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, Vaishampayan UN, Drabkin HA, George S, Logan TF, Margolin KA, Plimack ER, Lambert AM, Waxman IM, Hammers HJ. Nivolumab for Metastatic Renal Cell Carcinoma: Results of a Randomized Phase II Trial. J Clin Oncol 2015; 33:1430-7. [PMID: 25452452 PMCID: PMC4806782 DOI: 10.1200/jco.2014.59.0703] [Citation(s) in RCA: 841] [Impact Index Per Article: 84.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Nivolumab is a fully human immunoglobulin G4 programmed death-1 immune checkpoint inhibitor antibody that restores T-cell immune activity. This phase II trial assessed the antitumor activity, dose-response relationship, and safety of nivolumab in patients with metastatic renal cell carcinoma (mRCC). PATIENTS AND METHODS Patients with clear-cell mRCC previously treated with agents targeting the vascular endothelial growth factor pathway were randomly assigned (blinded ratio of 1:1:1) to nivolumab 0.3, 2, or 10 mg/kg intravenously once every 3 weeks. The primary objective was to evaluate the dose-response relationship as measured by progression-free survival (PFS); secondary end points included objective response rate (ORR), overall survival (OS), and safety. RESULTS A total of 168 patients were randomly assigned to the nivolumab 0.3- (n = 60), 2- (n = 54), and 10-mg/kg (n = 54) cohorts. One hundred eighteen patients (70%) had received more than one prior systemic regimen. Median PFS was 2.7, 4.0, and 4.2 months, respectively (P = .9). Respective ORRs were 20%, 22%, and 20%. Median OS was 18.2 months (80% CI, 16.2 to 24.0 months), 25.5 months (80% CI, 19.8 to 28.8 months), and 24.7 months (80% CI, 15.3 to 26.0 months), respectively. The most common treatment-related adverse event (AE) was fatigue (24%, 22%, and 35%, respectively). Nineteen patients (11%) experienced grade 3 to 4 treatment-related AEs. CONCLUSION Nivolumab demonstrated antitumor activity with a manageable safety profile across the three doses studied in mRCC. No dose-response relationship was detected as measured by PFS. These efficacy and safety results in mRCC support study in the phase III setting.
Collapse
Affiliation(s)
- Robert J Motzer
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| | - Brian I Rini
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - David F McDermott
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Bruce G Redman
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Timothy M Kuzel
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Michael R Harrison
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Ulka N Vaishampayan
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Harry A Drabkin
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Saby George
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Theodore F Logan
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Kim A Margolin
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Elizabeth R Plimack
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Alexandre M Lambert
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Ian M Waxman
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Hans J Hammers
- Robert J. Motzer, Memorial Sloan-Kettering Cancer Center, New York; Saby George, Roswell Park Cancer Institute, Buffalo, NY; Brian I. Rini, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; David F. McDermott, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA; Bruce G. Redman, University of Michigan Comprehensive Cancer Center, Ann Arbor; Ulka N. Vaishampayan, Karmanos Cancer Institute, Wayne State University, Detroit, MI; Timothy M. Kuzel, Northwestern University Feinberg School of Medicine, Chicago, IL; Michael R. Harrison, Duke University Medical Center, Durham, NC; Harry A. Drabkin, Medical University of South Carolina, Charleston, SC; Theodore F. Logan, Indiana University Simon Cancer Center, Indianapolis, IN; Kim A. Margolin, Stanford University, Stanford, CA; Elizabeth R. Plimack, Fox Chase Cancer Center, Philadelphia, PA; Alexandre M. Lambert, Bristol-Myers Squibb, Braine-l'Alleud, Belgium; Ian M. Waxman, Bristol-Myers Squibb, Princeton, NJ; and Hans J. Hammers, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
18
|
Unverzagt S, Moldenhauer I, Coppin C, Greco F, Seliger B. Immunotherapy for metastatic renal cell carcinoma. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2015. [DOI: 10.1002/14651858.cd011673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
19
|
Haddad AQ, Margulis V. Tumour and patient factors in renal cell carcinoma-towards personalized therapy. Nat Rev Urol 2015; 12:253-62. [PMID: 25868564 DOI: 10.1038/nrurol.2015.71] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Renal cell carcinoma (RCC) comprises a heterogeneous group of histologically and molecularly distinct tumour subtypes. Current targeted therapies have improved survival in patients with advanced disease but complete response occurs rarely, if at all. The genomic characterization of RCC is central to the development of novel targeted therapies. Large-scale studies employing multiple 'omics' platforms have led to the identification of key driver genes and commonly altered pathways. Specific molecular alterations and signatures that correlate with tumour phenotype and clinical outcome have been identified and can be harnessed for patient management and counselling. RCC seems to be a remarkably diverse malignancy with significant intratumour and intertumour genetic heterogeneity. The tumour microenvironment is increasingly recognized as a vital regulator of RCC tumour biology. Patient factors, including immune response and drug metabolism, vary widely, which can lead to widely divergent responses to drug therapy. Intratumour heterogeneity poses a significant challenge to the development of personalized therapies in RCC as a single biopsy might not accurately represent the clonal population ultimately responsible for aggressive biologic behaviour. On the other hand, the diversity of genomic alterations in RCC could also afford opportunities for targeting unique pathways based on analysis of an individual tumour's molecular composition.
Collapse
Affiliation(s)
- Ahmed Q Haddad
- Department of Urology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
20
|
Bedke J, Kruck S, Gakis G, Stenzl A, Goebell PJ. Checkpoint modulation--A new way to direct the immune system against renal cell carcinoma. Hum Vaccin Immunother 2015; 11:1201-8. [PMID: 25912622 PMCID: PMC4514323 DOI: 10.1080/21645515.2015.1016657] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/06/2015] [Accepted: 01/19/2015] [Indexed: 12/31/2022] Open
Abstract
The introduction of targeted therapies like the tyrosine kinase (TKI) and mammalian target of rapamycin (mTOR) inhibitors has improved patients' survival in general. Nevertheless the prognosis remains limited. Therapies with a new mode of action are urgently warranted, especially those who would provoke long-term responders or long-lasting complete remissions as observed with unspecific immunotherapy with the cytokines interleukin-2 and interferon-α. In the recent years a deeper understanding of the underlying immunology of T cell activation led to the development of checkpoint inhibitors, which are mainly monocloncal antibodies and which enhances the presence of the co-stimulatory signals needed for T cell activation or priming. This review discusses the clinical data and ongoing studies available for the inhibition of the PD-1 (CD279) and CTLA-4 (CD152) axis in mRCC. In addition, potential future immunological targets are discussed. This approach of T-cell activation or re-activation by immunological checkpoint inhibition holds the inherent promise to directly affect the tumor cell and thereby to potentially cure a subset of patients with mRCC.
Collapse
Affiliation(s)
- Jens Bedke
- Department of Urology; Eberhard Karls University; Tübingen, Germany
- German Cancer Consortium (DKTK); Partnerstandort Tübingen; German Cancer Research Center (DKFZ); Heidelberg, Germany
| | - Stephan Kruck
- Department of Urology; Eberhard Karls University; Tübingen, Germany
| | - Georgios Gakis
- Department of Urology; Eberhard Karls University; Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology; Eberhard Karls University; Tübingen, Germany
- German Cancer Consortium (DKTK); Partnerstandort Tübingen; German Cancer Research Center (DKFZ); Heidelberg, Germany
| | - Peter J Goebell
- Deptartment of Urology; Friedrich-Alexander University; Erlangen, Germany
| |
Collapse
|
21
|
Sharma RK, Yolcu ES, Shirwan H. SA-4-1BBL as a novel adjuvant for the development of therapeutic cancer vaccines. Expert Rev Vaccines 2014; 13:387-98. [PMID: 24521311 DOI: 10.1586/14760584.2014.880340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor associated antigen (TAA)-based therapeutic vaccines have great potential as a safe, practical, and cost-efficient alternative to standard treatments for cancer. Clinical efficacy of TAA-based vaccines, however, has yet to be realized and will require adjuvants with pleiotropic functions on immune cells. Such adjuvants need not only to generate/boost T cell responses, but also reverse intrinsic/extrinsic tumor immune evasion mechanisms for therapeutic efficacy. This review focuses on a novel agonistic ligand, SA-4-1BBL, for 4-1BB costimulatory receptor as an adjuvant of choice because of its ability to: i) serve as a vehicle to deliver TAAs to dendritic cells (DCs) for antigen uptake and cross-presentation to CD8(+) T cells; ii) augment adaptive Th1 and innate immune responses; and iii) overcome various immune evasion mechanisms, cumulatively translating into therapeutic efficacy in preclinical tumor models.
Collapse
Affiliation(s)
- Rajesh K Sharma
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | | | | |
Collapse
|
22
|
Rausch S, Kruck S, Stenzl A, Bedke J. IMA901 for metastatic renal cell carcinoma in the context of new approaches to immunotherapy. Future Oncol 2014; 10:937-48. [DOI: 10.2217/fon.14.61] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
ABSTRACT: The promising option of immunotherapy for metastatic renal cell carcinoma has evolved from rather unspecific approaches to a specific activation of an anti-tumor T-cell response. The latest step is a synthetic peptide vaccine called IMA901, which demonstrated a clear association between a provoked T-cell response and a prolonged overall survival. The results of IMA901 for the treatment of metastatic renal cell carcinoma are discussed together with new approaches to immunotherapy, such as local and systemic immunomodulation with adjuvants, checkpoint inhibitors, classical chemotherapeutics, such as cyclophosphamide or tyrosine kinase inhibitors. The capability of theses substances to modulate leukocytes subsets, such as myeloid-derived suppressor cells, Tregs or Th17 cells, are outlined together with the possibility to combine them with tumor vaccination strategies to achieve a higher cancer specificity and immunogenicity.
Collapse
Affiliation(s)
- Steffen Rausch
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
| | - Stephan Kruck
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jens Bedke
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
León L, García-Figueiras R, García-Figueras R, Suárez C, Arjonilla A, Puente J, Vargas B, Méndez Vidal MJ, Sebastiá C. Recommendations for the clinical and radiological evaluation of response to treatment in metastatic renal cell cancer. Target Oncol 2013; 9:9-24. [PMID: 24338498 DOI: 10.1007/s11523-013-0304-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 11/28/2013] [Indexed: 12/21/2022]
Abstract
The evaluation of response to treatment is a critical step for determining the effectiveness of oncology drugs. Targeted therapies such as tyrosine kinase inhibitors and mammalian target of rapamycin inhibitors are active drugs in patients with metastatic renal cell carcinoma (mRCC). However, treatment with this type of drugs may not result in significant reductions in tumor size, so standard evaluation criteria based on tumor size, such as Response Evaluation Criteria in Solid Tumors (RECIST), may be inappropriate for evaluating response to treatment in patients with mRCC. In fact, targeted therapies apparently yield low response rates that do not reflect increased disease control they may cause and, consequently, the benefit in terms of time to progression. To improve the clinical and radiological evaluation of response to treatment in patients with mRCC treated with targeted drugs, a group of 32 experts in this field have reviewed different aspects related to this issue and have put together a series of recommendations with the intention of providing guidance to clinicians on this matter.
Collapse
Affiliation(s)
- Luís León
- Medical Oncology Department, Complejo Hospitalario Universitario de Santiago, A Coruña, Spain,
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Seeliger B, Callari C, Diana M, Mutter D, Marescaux J. Polypoid Gallbladder Lesion in the Context of Renal Cell Carcinoma: Is Laparoscopic Cholecystectomy a Reasonable Option? J Investig Med High Impact Case Rep 2013; 1:2324709613499008. [PMID: 26425579 PMCID: PMC4586817 DOI: 10.1177/2324709613499008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Introduction. The only curative therapeutic approach for renal cell carcinoma (RCC) is surgery. Laparoscopic surgery for RCC has become an established surgical procedure with equivalent cancer-free survival rate, following the same surgical oncological principles as open surgery. Metastatic RCC of the gallbladder is a rare phenomenon. Hence, there are few reports regarding their management. Case Presentation. We report 2 cases of gallbladder metastasis from clear cell RCC treated by laparoscopic cholecystectomy. The first case was that of a 44-year-old male patient who underwent palliative cholecystectomy, the second case was that of an 83-year-old female patient who is doing well 55 months after surgery without evidence of disease recurrence. Conclusion. The outcome allows us to demonstrate the interest of surgical resection of RCC metastases in the gallbladder by laparoscopic cholecystectomy, respecting surgical oncological principles. Laparoscopic resection of an uncommon gallbladder metastasis can provide long-term favorable outcome.
Collapse
Affiliation(s)
| | | | - Michele Diana
- University Hospital of Strasbourg, Strasbourg, France
| | - Didier Mutter
- University Hospital of Strasbourg, Strasbourg, France
| | | |
Collapse
|