1
|
Meng L, Collier KA, Wang P, Li Z, Monk P, Mortazavi A, Hu Z, Spakowicz D, Zheng L, Yang Y. Emerging Immunotherapy Approaches for Advanced Clear Cell Renal Cell Carcinoma. Cells 2023; 13:34. [PMID: 38201238 PMCID: PMC10777977 DOI: 10.3390/cells13010034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The most common subtype of renal cell carcinoma is clear cell renal cell carcinoma (ccRCC). While localized ccRCC can be cured with surgery, metastatic disease has a poor prognosis. Recently, immunotherapy has emerged as a promising approach for advanced ccRCC. This review provides a comprehensive overview of the evolving immunotherapeutic landscape for metastatic ccRCC. Immune checkpoint inhibitors (ICIs) like PD-1/PD-L1 and CTLA-4 inhibitors have demonstrated clinical efficacy as monotherapies and in combination regimens. Combination immunotherapies pairing ICIs with antiangiogenic agents, other immunomodulators, or novel therapeutic platforms such as bispecific antibodies and chimeric antigen receptor (CAR) T-cell therapy are areas of active research. Beyond the checkpoint blockade, additional modalities including therapeutic vaccines, cytokines, and oncolytic viruses are also being explored for ccRCC. This review discusses the mechanisms, major clinical trials, challenges, and future directions for these emerging immunotherapies. While current strategies have shown promise in improving patient outcomes, continued research is critical for expanding and optimizing immunotherapy approaches for advanced ccRCC. Realizing the full potential of immunotherapy will require elucidating mechanisms of response and resistance, developing predictive biomarkers, and rationally designing combination therapeutic regimens tailored to individual patients. Advances in immunotherapy carry immense promise for transforming the management of metastatic ccRCC.
Collapse
Affiliation(s)
- Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
| | - Katharine A. Collier
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
| | - Peng Wang
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
| | - Zihai Li
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Paul Monk
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
| | - Zhiwei Hu
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
| | - Daniel Spakowicz
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Linghua Zheng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yuanquan Yang
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (K.A.C.); (P.W.); (Z.L.); (P.M.); (A.M.); (D.S.); (L.Z.)
| |
Collapse
|
2
|
Yao C, Zhang T, Wu T, Brugarolas J. Facts and Hopes for Immunotherapy in Renal Cell Carcinoma. Clin Cancer Res 2022; 28:5013-5020. [PMID: 35819272 PMCID: PMC9835201 DOI: 10.1158/1078-0432.ccr-21-2372] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/16/2022] [Accepted: 06/24/2022] [Indexed: 01/24/2023]
Abstract
Immunotherapy has made a significant impact in many tumors, including renal cell carcinoma (RCC). RCC has been known to be immunoresponsive since the cytokine era of IFNα and IL2, but only a small number of patients had durable clinical benefit. Since then, discoveries of key tumor drivers, as well as an understanding of the contribution of angiogenesis and the tumor microenvironment (TME), has led to advances in drug development, ultimately transforming patient outcomes. Combinations of anti-angiogenic agents with immune checkpoint inhibitors are now standard of care. Current challenges include patient selection for immunotherapy combinations, resistance acquisition, and optimally sequencing therapies. Further discoveries about RCC biology, the TME, and resistance mechanisms will likely pave the way for the next generation of therapies.
Collapse
Affiliation(s)
- Chen Yao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tian Zhang
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tuoqi Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
3
|
Zatovicova M, Kajanova I, Barathova M, Takacova M, Labudova M, Csaderova L, Jelenska L, Svastova E, Pastorekova S, Harris AL, Pastorek J. Novel humanized monoclonal antibodies for targeting hypoxic human tumors via two distinct extracellular domains of carbonic anhydrase IX. Cancer Metab 2022; 10:3. [PMID: 35109923 PMCID: PMC8811981 DOI: 10.1186/s40170-022-00279-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Hypoxia in the tumor microenvironment (TME) is often the main factor in the cancer progression. Moreover, low levels of oxygen in tumor tissue may signal that the first- or second-line therapy will not be successful. This knowledge triggers the inevitable search for different kinds of treatment that will successfully cure aggressive tumors. Due to its exclusive expression on cancer cells, carbonic anhydrase IX belongs to the group of the most precise targets in hypoxic tumors. CA IX possesses several exceptional qualities that predetermine its crucial role in targeted therapy. Its expression on the cell membrane makes it an easily accessible target, while its absence in healthy corresponding tissues makes the treatment practically harmless. The presence of CA IX in solid tumors causes an acidic environment that may lead to the failure of standard therapy. METHODS Parental mouse hybridomas (IV/18 and VII/20) were humanized to antibodies which were subsequently named CA9hu-1 and CA9hu-2. From each hybridoma, we obtained 25 clones. Each clone was tested for antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activity, affinity, extracellular pH measurement, multicellular aggregation analysis, and real-time monitoring of invasion with the xCELLigence system. RESULTS Based on the results from in vivo experiments, we have selected mouse monoclonal antibodies VII/20 and IV/18. The first one is directed at the conformational epitope of the catalytic domain, internalizes after binding to the antigen, and halts tumor growth while blocking extracellular acidification. The second targets the sequential epitope of the proteo-glycan domain, does not internalize, and is able to block the attachment of cancer cells to the matrix preventing metastasis formation. In vitro experiments prove that humanized versions of the parental murine antibodies, CA9hu-1 and CA9hu-2, have preserved these characteristics. They can reverse the failure of standard therapy as a result of an acidic environment by modulating the TME, and both are able to induce an immune response and have high affinity, as well as ADCC and CDC activity. CONCLUSION CA9hu-1 and CA9hu-2 are the very first humanized antibodies against CA IX that are likely to become suitable therapies for hypoxic tumors. These antibodies can be applied in the treatment therapy of primary tumors and suppression of metastases formation.
Collapse
Affiliation(s)
- Miriam Zatovicova
- MABPRO, a.s., Dubravska cesta 2, 841 04, Bratislava, Slovakia
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Ivana Kajanova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Monika Barathova
- MABPRO, a.s., Dubravska cesta 2, 841 04, Bratislava, Slovakia
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Martina Takacova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Martina Labudova
- MABPRO, a.s., Dubravska cesta 2, 841 04, Bratislava, Slovakia
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Lucia Csaderova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Lenka Jelenska
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Eliska Svastova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Silvia Pastorekova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Adrian L Harris
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | | |
Collapse
|
4
|
Abstract
Harnessing the power of the human immune system to treat cancer is the essence of immunotherapy. Monoclonal antibodies engage the innate immune system to destroy targeted cells. For the last 30years, antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity have been the main mechanisms of anti-tumor action of unconjugated antibody drugs. Efforts to exploit the potentials of other immune cells, in particular T cells, culminated in the recent approval of two T cell engaging bispecific antibody (T-BsAb) drugs, thereby stimulating new efforts to accelerate similar platforms through preclinical and clinical trials. In this review, we have compiled the worldwide effort in exploring T cell engaging bispecific antibodies. Our special emphasis is on the lessons learned, with the hope to derive insights in this fast evolving field with tremendous clinical potential.
Collapse
Affiliation(s)
- Z Wu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - N V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
5
|
Pastorek J, Pastorekova S. Hypoxia-induced carbonic anhydrase IX as a target for cancer therapy: from biology to clinical use. Semin Cancer Biol 2014; 31:52-64. [PMID: 25117006 DOI: 10.1016/j.semcancer.2014.08.002] [Citation(s) in RCA: 242] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 07/30/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment includes a complicated network of physiological gradients contributing to plasticity of tumor cells and heterogeneity of tumor tissue. Hypoxia is a key component generating intratumoral oxygen gradients, which affect the cellular expression program and lead to therapy resistance and increased metastatic propensity of weakly oxygenated cell subpopulations. One of the adaptive responses of tumor cells to hypoxia involves the increased expression and functional activation of carbonic anhydrase IX (CA IX), a cancer-related cell surface enzyme catalyzing the reversible conversion of carbon dioxide to bicarbonate ion and proton. Via its catalytic activity, CA IX participates in regulation of intracellular and extracellular pH perturbations that result from hypoxia-induced changes in cellular metabolism producing excess of acid. Through the ability to regulate pH, CA IX also facilitates cell migration and invasion. In addition, CA IX has non-catalytic function in cell adhesion and spreading. Thus, CA IX endows tumor cells with survival advantages in hypoxia/acidosis and confers an increased ability to migrate, invade and metastasize. Accordingly, CA IX is expressed in a broad range of tumors, where it is associated with prognosis and therapy outcome. Its expression pattern and functional implications in tumor biology make CA IX a promising therapeutic target, which can be hit either by immunotherapy with monoclonal antibodies or with compounds inhibiting its enzyme activity. The first strategy has already reached the clinical trials, whereas the second one is still in preclinical testing. Both strategies indicate that CA IX can become a clinically useful anticancer target, but urge further efforts toward better selection of patients for immunotherapy and deeper understanding of tumor types, clinical situations and synthetic lethality interactions with other treatment approaches.
Collapse
Affiliation(s)
- Jaromir Pastorek
- Department of Molecular Medicine, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Silvia Pastorekova
- Department of Molecular Medicine, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia; Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
| |
Collapse
|
6
|
Oberneder R, Siebels M, Hofstetter A. Adjuvante Therapie des Nierenzellkarzinoms. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/s001310050213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
7
|
|
8
|
Liu J, Fang L, Cheng Q, Li L, Su C, Zhang B, Pei D, Yang J, Li W, Zheng J. Effects of G250 promoter controlled conditionally replicative adenovirus expressing Ki67-siRNA on renal cancer cell. Cancer Sci 2012; 103:1880-8. [PMID: 22775978 DOI: 10.1111/j.1349-7006.2012.02380.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 06/20/2012] [Accepted: 06/22/2012] [Indexed: 12/27/2022] Open
Abstract
Replication-competent adenovirus (RCAd) has been used extensively in cancer gene therapy, and tumor-selection is critical for the use of replication-competent adenovirus. Here we investigated the anti-tumor characterization of oncolytic virus, whose E1A gene is under the control of a renal cell carcinoma specific promoter - the G250 promoter. The constructed oncolytic virus G250-Ki67 is armed with transgene of Ki67-siRNA, and G250-ZD55-Ki67 also with E1B-55 KD deleted. The tumor-specific expression of E1A and Ki67 was demonstrated by Western blot and immunohistochemistry staining, and the tumor-specific cytotoxicity was assessed by crystal violet staining and cell viability assays. The G250-Ki67 and G250-ZD55-Ki67 adenoviruses could express E1A protein in 786-O and OSRC cell lines but not in ACHN and HK-2 cell lines. The expression of Ki67 gene in 786-O and OSRC cell lines were suppressed by these adenoviruses. The cytotoxic effects induced by G250-ZD55-Ki67 and G250-Ki67 were more obvious on the 786-O cell lines than on the OSRC cell lines. Each group of adenoviruses could inhibit the proliferation of the 786-O cells and OSRC cells. However, the effects induced by G250-ZD55-Ki67 and G250-Ki67 on 786-O cells were stronger than on OSRC cells. Moreover, G250-ZD55-Ki67 had enhanced antitumor activities in these renal cancer cells compared with G250-Ki67. G250 promoter-derived CRAds carrying Ki67-siRNA could highly amplify and express Ki67-siRNA in renal cancer cells with expression of G250 antigen, inhibit renal cancer cells proliferation and induce apoptosis. These results demonstrated that the G250-specific oncolytic adenovirus expressing Ki67-siRNA is applicable for human renal clear cell cancer therapy.
Collapse
Affiliation(s)
- Junjie Liu
- Laboratory of Urology, Affiliated Hospital of Xuzhou Medical College, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
Bispecific antibodies (BiAbs) offer a unique opportunity to redirect immune effector cells to kill cancer cells. BiAbs combine the benefits of different binding specificities of two monoclonal antibodies (mAbs) into a single construct. This unique feature of BiAbs enables approaches that are not possible with single mAbs. Advances in antibody engineering and antigen profiling of malignant cells have led to the development of a number of BiAb formats and their combinations for redirecting effector cells to tumor targets. There have been significant advances in the design and application of BiAbs for intravenous and local injection.The initial barrier of cytokine storm has been partially overcome by more recent constructs that have improved clinical effectiveness without dose-limiting toxicities. Since the recent revival of BiAbs, there has been multiple, ongoing, phase I/II and III trials, and some promising clinical outcomes have been reported in completed clinical studies. This review focuses on arming T cells with BiAbs to create the 'poor man's cytotoxic lymphocyte'.
Collapse
Affiliation(s)
- Lawrence G Lum
- Department of Oncology, Wayne State University and Barbara Ann Karmanos Cancer Center, Detroit, MI, USA
| | | |
Collapse
|
10
|
Lum LG, Al-Kadhimi Z. Development and prospects for bispecific antibody-based therapeutics in cancer and other applications. Expert Opin Drug Discov 2008; 3:1081-97. [PMID: 23506181 DOI: 10.1517/17460441.3.9.1081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Progress in molecular biology for the production of bispecific antibodies (BiAbs) and the expanding knowledge on receptors on malignant and normal target cells provide the basis for developing new strategies using antibody- and/or receptor-based platform technology for the treatment of cancer and other diseases. OBJECTIVE This review provides a preclinical and clinical perspective on application of bispecific antibodies for the treatment of solid tumors, hematologic malignancies and other diseases. METHODS This review focuses on BiAb-based immunotherapy, clinical trials, alternative strategies, the challenges of technology and future applications. RESULTS/CONCLUSION The successful application of a particular BiAb will depend on a thorough evaluation of the expected functional application and thoughtful engineering of structure, affinity and number of binding sites based on the desired function.
Collapse
Affiliation(s)
- Lawrence G Lum
- Professor of Medicine and Professor of Immunology and Microbiology, Scientific Director of BMT and Immunotherapy Program, Wayne State University, Member Barbara Ann Karmanos Cancer Institute, Hudson-Webber Cancer Research Center, 4100 John R., 7th Floor, Rm 740.1, Detroit, Michigan, MI 48201, USA +1 313 576 8326 ; +1 313 576 8939 ;
| | | |
Collapse
|
11
|
Collins S, McKiernan J, Landman J. Update on the epidemiology and biology of renal cortical neoplasms. J Endourol 2007; 20:975-85. [PMID: 17206887 DOI: 10.1089/end.2006.20.975] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
A new era is developing in the understanding of the diagnosis, classification, and management of renal-cell carcinoma (RCC). Historically, RCC has been divided into subtypes on the basis of the histopathologic findings alone. Now, genetic alterations, nuclear characteristics, and clinical criteria are routinely incorporated into the classification. The greater use of axial imaging that began in the 1980s dramatically increased the incidence of RCC, but there has not been a decrease in the percentage of cases that are metastatic. Nevertheless, many incidental lesions prove to be benign, so there is renewed enthusiasm for biopsy before treatment is selected. Genetic conditions associated with RCC, such as Von Hippel Lindau and Birt-Hogg-Dube syndromes, along with genetic analyses of tumors, have provided considerable insight into the pathogenesis of these lesions. Renal-cell carcinoma is resistant to chemotherapy, and high-dose interleukin-2 is the only regimen currently approved by the Food and Drug Administration for the treatment of advanced RCC. Stem cell transplantation is an evolving therapy. The vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), and transforming growth factor-alpha pathways are promising targets for medical therapy of RCC. Bevacizumab, a monoclonal antibody that acts as a competitive blocker of the VEGF receptor; sorafenib, an oral well-tolerated tyrosine kinase inhibitor that blocks the intracellular second-messenger system associated with the VEGF receptor; sunitinib, a multitarget inhibitor of kinases associated with the VEGF and PDGF receptors; temsirolimus (CCI-779), a kinase blocker that inhibits the mammalian target of rapamycin pathway; and erlotinib, an inhibitor of the tyrosine kinases associated with the EGF receptor, have shown promise. Combinations of the above therapies and cytokines also are being investigated, as there may be synergistic effects.
Collapse
Affiliation(s)
- Sean Collins
- Department of Urology, Columbia University, New York, New York, USA
| | | | | |
Collapse
|
12
|
Pastorekova S, Parkkila S, Zavada J. Tumor-associated carbonic anhydrases and their clinical significance. Adv Clin Chem 2006. [PMID: 17131627 DOI: 10.1016/s0065-2423(06)42005-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Carbonic anhydrases (CAs) are physiologically important enzymes that catalyze a reversible conversion of carbon dioxide to bicarbonate and participate in ion transport and pH control. Two human isoenzymes, CA IX and CA XII, are overexpressed in cancer and contribute to tumor physiology. Particularly CA IX is confined to only few normal tissues but is ectopically induced in many tumor types mainly due to its strong transcriptional activation by hypoxia accomplished via HIF-1 transcription factor. Therefore, CA IX can serve as a surrogate marker of hypoxia and a prognostic indicator. CA IX appears implicated in cell adhesion and in balance of pH disturbances caused by tumor metabolism. Both tumor-related expression pattern and functional involvement in tumor progression make it a suitable target for anticancer treatment. Here we summarize a current knowledge on CA IX and CA XII, and discuss possibilities of their exploitation for cancer detection, diagnostics, and therapy.
Collapse
Affiliation(s)
- Silvia Pastorekova
- Centre of Molecular Medicine, Institute of Virology, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovak Republic.
| | | | | |
Collapse
|
13
|
Reusch U, Sundaram M, Davol PA, Olson SD, Davis JB, Demel K, Nissim J, Rathore R, Liu PY, Lum LG. Anti-CD3 x anti-epidermal growth factor receptor (EGFR) bispecific antibody redirects T-cell cytolytic activity to EGFR-positive cancers in vitro and in an animal model. Clin Cancer Res 2006; 12:183-90. [PMID: 16397041 DOI: 10.1158/1078-0432.ccr-05-1855] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Targeting epidermal growth factor receptor (EGFR) overexpressed by many epithelial-derived cancer cells with anti-EGFR monoclonal antibodies (mAb) inhibits their growth. A limited number of clinical responses in patients treated with the anti-EGFR mAb, (cetuximab), may reflect variability in EGFR type or signaling in neoplastic cells. This study combines EGFR-targeting with the non-MHC-restricted cytotoxicity of anti-CD3 activated T cells (ATC) to enhance receptor-directed cytotoxicity. EXPERIMENTAL DESIGN ATC from normal and patient donors were expanded ex vivo. Specific cytolytic activity of ATC armed with anti-CD3 x anti-EGFR (EGFRBi) against EGFR-expressing cancer cells derived from lung, pancreas, colon, prostate, brain, skin, or EGFR-negative breast cancer cells was evaluated in (51)Cr release assays. In vivo studies comparing tumor growth delay induced by EGFRBi-armed ATCs or cetuximab were done in severe combined immunodeficient/Beige mice (SCID-Beige) bearing COLO 356/FG pancreatic and LS174T colorectal tumors. RESULTS At effector/target ratios from 3.125 to 50, both EGFRBi-armed normal and patient ATC were significantly more cytotoxic, by 23% to 79%, against EGFR-positive cells over ATC, cetuximab, anti-CD3 alone, or ATC armed with irrelevant BiAb directed at CD20. EGFRBi-armed ATC also secreted significantly higher levels of some T(H1)/T(H2) cytokines compared with ATC alone. In mice, i.v. infusions of EGFRBi-armed ATC (0.001 mg equivalent/infusion) were equally effective as cetuximab (1 mg/infusion) alone for significantly delaying growth of established COLO 356/FG but not LS174T tumors compared with mice that received ATC alone or vehicle (P < 0.001). CONCLUSIONS Combining EGFR antibody targeting with T cell-mediated cytotoxicity may overcome some limitations associated with EGFR-targeting when using cetuximab alone.
Collapse
Affiliation(s)
- Ursula Reusch
- Cancer Immunotherapy Program, Adele R. Decof Cancer Center, Providence, Rhode Island, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Affiliation(s)
- Herbert T Cohen
- Renal and Hematology-Oncology Sections, Department of Medicine, Boston University School of Medicine, Evans Biomedical Research Center, Boston, MA 02118, USA.
| | | |
Collapse
|
15
|
Abstract
Renal cell carcinoma (RCC) still represents a therapeutic challenge when patients have advanced or metastatic disease. Treatment using IL-2 and IFN-alpha continues to be the standard of care in patients who are able to tolerate such regimens. Targeted therapy may become the first-line treatment for patients resistant or intolerant to cytokines as new emerging drugs continue to be investigated. Understanding the genetic abnormalities related to the development of RCC (e.g., VHL gene abnormalities) and identifying molecular targets (e.g., epidermal growth factor, vascular endothelial growth factor and carbonic anhydrase IX) are playing a major role in the emergence of these novel agents for the treatment of this malignancy. Overall, these drugs are better tolerated and more acceptable to use by patients than the traditional cytokine-based regimens. The use of oral drugs to treat various malignancies including RCC seems to be the new paradigm of the future. Further understanding of their mechanisms of action and confirmation of their benefits on the clinical outcome is needed.
Collapse
Affiliation(s)
- Philip E Shaheen
- Cleveland Clinic Foundation, Experimental Therapeutics Program, Taussig Cancer Center, R33, Cleveland, OH 44195, USA
| | | |
Collapse
|
16
|
Lum LG, Davol PA. Retargeting T cells and immune effector cells with bispecific antibodies. CANCER CHEMOTHERAPY AND BIOLOGICAL RESPONSE MODIFIERS ANNUAL 2005; 22:273-91. [PMID: 16110617 DOI: 10.1016/s0921-4410(04)22013-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The development of BiAbs for therapeutic applications in cancer shows promise. As our understanding of effector cell receptor biology for triggering of cytotoxic functions improves and the behavior of TAA and the targeting antibody engagement is elucidated, customized BiAb reagents can be engineered to optimize in vivo or ex vivo arming of T cells for targeting tumors. Additionally, other variables that require consideration in the equation for successful T cell immunotherapy include: the type of effector cells, their state of activation, the type of effector receptor being activated or tareeted. the presence of Tregs, the affinity of the anti-effector cell antibody and the anti-TAA antibody, the type of BiAb (mouse, humanized, or human), the number of binding sites for the T cells or TAA, the presence or absence of decoy antigen, whether the TAA modulates after being engaged by antibody, the type of tumor, the tumor burden, and last, but not least, the amount of 'immunologic' space available for the adoptively transferred cells to expand and function.
Collapse
Affiliation(s)
- Lawrence G Lum
- Immunotherapy Program, Adele R. Deof Cancer Center, Roger Williams Hospital, Providence, RI 02908, USA.
| | | |
Collapse
|
17
|
Abstract
This article reviews the recent progress in the search for new treatments for renal cell cancer (RCC), based on a variety of preclinical models or strategies. Some recent clinical trials addressing migrating treatments from other cancers onto RCC and novel agents are discussed, as well as the molecular targets for some of the novel agents. Drugs oriented to histologically definable RCC features, such as the G250 antigen, and the receptor tyrosine kinases, such as epidermal growth factor receptor, are reviewed. Drugs aimed at antiangiogenesis and perturbing features of the cell cycle are also mentioned, including preclinical and empirical experience. Molecular techniques in the study of von Hippel Lindau-related pathways and mRNA expression analyses are cited. Within the immune model of therapy, progress in the application of immune-related drugs including older cytokines (IL-2, IFN-alpha) and of newer cytokine-variant and other cytokines are discussed. Finally, cell-based therapies such as lymphocyte infusions, tumour-cell vaccines, dendritic cell vaccines and allogeneic mini-transplant are outlined. Although high percentage improvements in outcomes for metastatic RCC are not yet realised, the many fronts for scientific and clinical advances form some basis for optimism in the coming years.
Collapse
Affiliation(s)
- Mayer Fishman
- H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, MCC 4035, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | | |
Collapse
|
18
|
Palapattu GS, Reiter RE. Monoclonal antibody therapy for genitourinary oncology: promise for the future. J Urol 2002; 168:2615-23. [PMID: 12441996 DOI: 10.1016/s0022-5347(05)64230-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Metastatic or recurrent cancer continues to be the bane of the urological oncologist. Despite recent improvements in therapeutic strategies and outcomes for clinically localized disease overall survival in patients with the majority of metastatic and recurrent genitourinary malignancies remains relatively unchanged. Modern advances in the field of immunotherapy hold the promise of providing the clinical urologist/oncologist with new tools to fight urological cancer. In this review we discuss the various mAb based strategies currently under investigation for urological oncology as well as the lessons learned from similar approaches in other fields. MATERIALS AND METHODS We reviewed the literature on mAb based immunotherapy with a particular emphasis on target antigens, mAb design and potential applications in the field of urology. RESULTS Early trials with mAb therapy for solid tumor oncology met with limited success due to difficulty with mAb design and production, the development of host immunological responses against murine monoclonal antibodies (that is human anti-mouse antibodies), suboptimal target antigen selection, and poor monoclonal antibody pharmacokinetics and tumor tissue penetration. Recent advances in the fields of immunology and oncology have sought to circumvent these obstacles. Today several preliminary studies have shown the effectiveness and usefulness of mAb based strategies for urological oncology. CONCLUSIONS The field of mAb based immunotherapy continues to evolve. New discoveries in this burgeoning area of cancer therapy show promise for the future.
Collapse
|
19
|
|
20
|
EPIDERMAL GROWTH FACTOR RECEPTOR AND G250:. J Urol 2002. [DOI: 10.1097/00005392-200202000-00070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Abstract
Metastatic renal cell cancer remains a disease which is difficult to treat medically. Prognosis often depends more on intrinsic disease features than on treatment choices. In this review, we examine novel therapies and scientific directions surrounding the RCC treatment problem. Reports relating chromosomal aberrations and of comparative gene expression analyses relating to RCC, are reviewed briefly. The central role of the von Hippel Lindau protein in clear cell RCC pathogenesis is evident. The limited contribution of conventional cytotoxic chemotherapy is mentioned. Some clinically applied agents whose clinical results are highlighted include 5-FU, retinoids, thalidomide, razoxane and IL-12. Features of the pathophysiology of von Hippel Lindau protein are described, with attention to potential novel therapies targeting HIF-1alpha, VEGF, TGF-beta1 and TGF-alpha pathways. Immunotherapy is being explored in many angles. Most basic are cytokine therapies incorporating new IL-2 and IFN-alpha schedules. Newer cytokine-based drugs include pegylated forms and IL-12. Allogeneic mini-transplantation has generated much interest. Tumour-associated antigens are being used to direct therapy using both identified and non-identified epitopes. A variety of tumour-cell vaccine and dendritic-cell vaccine clinical approaches are discussed. Finally, nephrectomy for known metastatic disease has been demonstrated to be helpful in retrospective and now prospective trials. Resection of metastases is also discussed. We are optimistic that the further clinical development among these novel therapies will improve the outlook for metastatic RCC.
Collapse
Affiliation(s)
- M Fishman
- H Lee Moffitt Cancer Center & Research Institute, University of South Florida, Interdisciplinary Oncology Program, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | | | | |
Collapse
|
22
|
Abstract
Metastatic renal cell carcinoma remains a disease that is highly resistant to systemic therapy and difficult to treat. Numerous studies with many different treatment modalities have resulted in only minor advances. No single agent or combination therapy has consistently shown a response proportion of 20% or higher. Interleukin-2 and interferon-alpha-based therapies are most commonly used to treat advanced disease, demonstrating low but reproducible response proportions in the 10% to 20% range, with durable responses of 5% or less. In the last few years, randomized studies have for the first time demonstrated a survival advantage for patients receiving systemic therapy, although this advantage is marginal. A number of novel treatment strategies are being investigated with some encouraging early results. The identification of new agents with better antitumor activity remains a high priority in the treatment of advanced renal cell carcinoma.
Collapse
Affiliation(s)
- D M Nanus
- Division of Hematology and Medical Oncology, Departments of Medicine and Urology, Joan and Sanford I. Weill Medical College of Cornell University, 525 East 68th Street, B-1519, New York, NY 10021, USA.
| |
Collapse
|
23
|
De Jonge J, Heirman C, Veerman MD, Van Meirvenne S, Moser M, Leo O, Thielemans K. In Vivo Retargeting of T Cell Effector Function by Recombinant Bispecific Single Chain Fv (Anti-CD3 × Anti-Idiotype) Induces Long-Term Survival in the Murine BCL1 Lymphoma Model. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.3.1454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
As demonstrated in several preclinical models, bispecific Abs are attractive immunotherapeutic agents for tumor treatment. We have previously reported that a bacterially produced anti-CD3 × antitumor bispecific single chain variable fragment of Ab fragment (BsscFv), which is capable of retargeting CTLs toward BCL1 tumor cells, exhibits antitumor activity in vitro. To further facilitate BsscFv production, the coding sequence was subcloned in a eukaryotic expression vector and introduced into Chinese hamster ovary cells for large-scale production. In this report, we have determined the serum stability and the clearance rate from the circulation of BsscFv. Most important, we prove here the therapeutic value of BsscFv in the treatment of BCL1 lymphoma, a murine model for human non-Hodgkin’s lymphoma. Tumor-bearing mice that were treated with rscFv in combination with staphylococcal enterotoxin B superantigen, human rIL-2, or murine rIL-12 showed long-term survival, whereas untreated mice all died. This is the first report of the successful in vivo use of BsscFv as an immunotherapeutic agent. Furthermore, long-term survival was the result of complete tumor removal and was not due to the induction of dormancy.
Collapse
Affiliation(s)
- Jan De Jonge
- *Vrije Universiteit Brussel, Medical School, Laboratory of Physiology-Immunology, Brussels, Belgium; and
| | - Carlo Heirman
- *Vrije Universiteit Brussel, Medical School, Laboratory of Physiology-Immunology, Brussels, Belgium; and
| | - Marijke de Veerman
- *Vrije Universiteit Brussel, Medical School, Laboratory of Physiology-Immunology, Brussels, Belgium; and
| | - Sonja Van Meirvenne
- *Vrije Universiteit Brussel, Medical School, Laboratory of Physiology-Immunology, Brussels, Belgium; and
| | - Muriel Moser
- †Université Libre de Bruxelles, Laboratoire de Physiologie Animale, St-Genesius-Rode, Belgium
| | - Oberdan Leo
- †Université Libre de Bruxelles, Laboratoire de Physiologie Animale, St-Genesius-Rode, Belgium
| | - Kris Thielemans
- *Vrije Universiteit Brussel, Medical School, Laboratory of Physiology-Immunology, Brussels, Belgium; and
| |
Collapse
|
24
|
Abstract
Clinical research in the area of antibody-based tumor-targeted therapy has been driven for many years by the prospect of identifying cell surface antigens with sufficient restrictive tissue expression patterns to allow for the selective and specific accumulation of antibody in tumor tissue. Few, if any, such antibody-antigen systems have been identified which can effectively deliver a large fraction of an administered therapeutic agent to metastatic cancer. Despite this limitation, however, a greater understanding of the biological and physiological principles of tumor-targeted therapy has resulted in successful antibody-based therapy of lymphoma, colon cancer and breast cancer in recent clinical trials.
Collapse
Affiliation(s)
- A M Scott
- Ludwig Institute for Cancer Research, Melbourne Tumour Biology Branch, Austin, Australia.
| | | |
Collapse
|