1
|
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: 8.0] [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
|
2
|
Huang JJ, Hsieh JJ. The Therapeutic Landscape of Renal Cell Carcinoma: From the Dark Age to the Golden Age. Semin Nephrol 2021; 40:28-41. [PMID: 32130964 DOI: 10.1016/j.semnephrol.2019.12.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Oncologic treatments for renal cell carcinoma (RCC) have undergone a major revolution in the past 2 decades, moving away from the pre-2004 Dark Age during which interleukin 2 and interferon-α were the only therapeutic options and induced treatment responses in only 5% to 10% of patients with metastatic disease. The development of anti-angiogenic tyrosine kinase inhibitors against vascular endothelial growth factor receptor 2 and inhibitors of mammalian target of rapamycin complex 1 in 2005 introduced the Modern Age with better overall and progression-free survival and a greater number of patients (30%-40%) responding to and (∼80%) benefiting from these targeted therapeutic agents. The coming of age of the immuno-oncology era with the use of immune checkpoint inhibitors (ICIs) have ushered us into the Golden Age of metastatic RCC care, in which combined administrations of two ICIs (anti-programmed cell death protein 1/programmed death-ligand 1 and anti-cytotoxic T-lymphocyte-associated protein 4 or one tyrosine kinase inhibitor plus one ICI (anti-programmed cell death protein 1/programmed death-ligand 1) have recast the treatment landscape of clear cell RCC, the most common RCC subtype, with an approximately 60% response rate and an approximately 90% disease control rate that further improves metastatic RCC survival. Exciting clinical trials are in the pipeline investigating complementary/synergistic molecular mechanisms, based on studies investigating the biology, pathology, and genomics of renal carcinoma and the respective treatment outcome. This will enable us to enter the Diamond Age of precision medicine in which a specific treatment can be tailored to the specific biological and pathologic circumstance of an individual kidney tumor to offer more effective yet less toxic therapy.
Collapse
Affiliation(s)
- Jennifer J Huang
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
| | - James J Hsieh
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO.
| |
Collapse
|
3
|
Szeliga M. Thiadiazole derivatives as anticancer agents. Pharmacol Rep 2020; 72:1079-1100. [PMID: 32880874 PMCID: PMC7550299 DOI: 10.1007/s43440-020-00154-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023]
Abstract
In spite of substantial progress made toward understanding cancer pathogenesis, this disease remains one of the leading causes of mortality. Thus, there is an urgent need to develop novel, more effective anticancer therapeutics. Thiadiazole ring is a versatile scaffold widely studied in medicinal chemistry. Mesoionic character of this ring allows thiadiazole-containing compounds to cross cellular membrane and interact strongly with biological targets. Consequently, these compounds exert a broad spectrum of biological activities. This review presents the current state of knowledge on thiadiazole derivatives that demonstrate in vitro and/or in vivo efficacy across the cancer models with an emphasis on targets of action. The influence of the substituent on the compounds' activity is depicted. Furthermore, the results from clinical trials assessing thiadiazole-containing drugs in cancer patients are summarized.
Collapse
Affiliation(s)
- Monika Szeliga
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Str, 02-106, Warsaw, Poland.
| |
Collapse
|
4
|
2-Amino-1,3,4-thiadiazoles as prospective agents in trypanosomiasis and other parasitoses. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2020; 70:259-290. [PMID: 32074064 DOI: 10.2478/acph-2020-0031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 01/19/2023]
Abstract
Parasitic diseases are a serious public health problem affecting hundreds of millions of people worldwide. African trypanosomiasis, American trypanosomiasis, leishmaniasis, malaria and toxoplasmosis are the main parasitic infections caused by protozoan parasites with over one million deaths each year. Due to old medications and drug resistance worldwide, there is an urgent need for new antiparasitic drugs. 1,3,4-Thiadiazoles have been widely studied for medical applications. The chemical, physical and pharmacokinetic properties recommend 1,3,4-thiadiazole ring as a target in drug development. Many scientific papers report the antiparasitic potential of 2-amino-1,3,4-thiadiazoles. This review presents synthetic 2-amino-1,3,4-thiadiazoles exhibiting antitrypanosomal, antimalarial and antitoxoplasmal activities. Although there are insufficient results to state the quality of 2-amino-1,3,4-thiadiazoles as a new class of antiparasitic agents, many reported derivatives can be considered as lead compounds for drug synthesis and a promise for the future treatment of parasitosis and provide a valid strategy for the development of potent antiparasitic drugs.
Collapse
|
5
|
Prasad MG, Lakshmi CV, Katari NK, Pal M. Lemon Juice as a Biocatalyst Under Ultrasound Irradiation: Synthesis and Pharmacological Evaluation of 2-amino 1,3,4-thiadiazoles. Anticancer Agents Med Chem 2020; 20:1379-1386. [PMID: 32271700 DOI: 10.2174/1871520620666200409143513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/19/2020] [Accepted: 02/12/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The 2-amino 1,3,4-thiadiazole framework has attracted considerable interest because of its prevalence in compounds possessing a wide range of pharmacological properties including anticancer/antitumor activities. Though a number of methods have been reported for the synthesis of this class of compounds, some of them are not straightforward, inexpensive and environmentally friendly. OBJECTIVE To synthesize 2-amino-1,3,4-thiadiazole derivatives that could act as potential anticancer agents. METHODS The use of lemon juice as an inexpensive and readily available biocatalyst was explored in the synthesis of 2-amino 1,3,4-thiadiazole derivatives. Accordingly, a convenient method has been developed for the rapid synthesis of this class of compounds under a mild and non-hazardous reaction condition in good yields. The methodology involved the reaction of various acid hydrazides with TMSNCS in the presence of lemon juice in PEG-400 at room temperature (25-30ºC) under ultrasound irradiation. These compounds were assessed for their cytotoxic properties against two different metastatic breast cancer cell lines e.g., MDAMB-231 and MCF-7 and subsequently against SIRT1. RESULTS The 2-amino 1,3,4-thiadiazole derivatives 3a, 3i, 3j and 3l showed promising growth inhibition of MDAMB- 231 and MCF-7 cell lines and SIRT1 inhibition in vitro. Indeed, 3i was found to be a potent inhibitor of SIRT1. CONCLUSION An ultrasound-assisted method facilitated by lemon juice has been developed to synthesize 2-amino- 1,3,4-thiadiazole derivatives that could act as potential anticancer agents.
Collapse
Affiliation(s)
- Malavattu G Prasad
- Department of Chemistry, GITAM Deemed to be University, HTP Campus, Rudraram, Medak, Telangana-502 329, India
| | - Chapala V Lakshmi
- Department of Chemistry, GITAM Deemed to be University, HTP Campus, Rudraram, Medak, Telangana-502 329, India
| | - Naresh K Katari
- Department of Chemistry, GITAM Deemed to be University, HTP Campus, Rudraram, Medak, Telangana-502 329, India
| | - Manojit Pal
- Dr Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad-500046, India
| |
Collapse
|
6
|
Serban G. Future Prospects in the Treatment of Parasitic Diseases: 2-Amino-1,3,4-Thiadiazoles in Leishmaniasis. Molecules 2019; 24:E1557. [PMID: 31010226 PMCID: PMC6514673 DOI: 10.3390/molecules24081557] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/14/2019] [Accepted: 04/17/2019] [Indexed: 02/08/2023] Open
Abstract
Neglected tropical diseases affect the lives of a billion people worldwide. Among them, the parasitic infections caused by protozoan parasites of the Trypanosomatidae family have a huge impact on human health. Leishmaniasis, caused by Leishmania spp., is an endemic parasitic disease in over 88 countries and is closely associated with poverty. Although significant advances have been made in the treatment of leishmaniasis over the last decade, currently available chemotherapy is far from satisfactory. The lack of an approved vaccine, effective medication and significant drug resistance worldwide had led to considerable interest in discovering new, inexpensive, efficient and safe antileishmanial agents. 1,3,4-Thiadiazole rings are found in biologically active natural products and medicinally important synthetic compounds. The thiadiazole ring exhibits several specific properties: it is a bioisostere of pyrimidine or benzene rings with prevalence in biologically active compounds; the sulfur atom increases lipophilicity and combined with the mesoionic character of thiadiazoles imparts good oral absorption and good cell permeability, resulting in good bioavailability. This review presents synthetic 2-amino-1,3,4-thiadiazole derivatives with antileishmanial activity. Many reported derivatives can be considered as lead compounds for the synthesis of future agents as an alternative to the treatment of leishmaniasis.
Collapse
Affiliation(s)
- Georgeta Serban
- Pharmaceutical Chemistry Department, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga, 410028 Oradea, Romania.
| |
Collapse
|
7
|
Hoerner CR, Chen VJ, Fan AC. The 'Achilles Heel' of Metabolism in Renal Cell Carcinoma: Glutaminase Inhibition as a Rational Treatment Strategy. KIDNEY CANCER 2019; 3:15-29. [PMID: 30854496 PMCID: PMC6400133 DOI: 10.3233/kca-180043] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
An important hallmark of cancer is 'metabolic reprogramming' or the rewiring of cellular metabolism to support rapid cell proliferation [1-5]. Metabolic reprogramming through oncometabolite-mediated transformation or activation of oncogenes in renal cell carcinoma (RCC) globally impacts energy production as well as glucose and glutamine utilization in RCC cells, which can promote dependence on glutamine supply to support cell growth and proliferation [6, 7]. Novel inhibitors of glutaminase, a key enzyme in glutamine metabolism, target glutamine addiction as a viable treatment strategy in metastatic RCC (mRCC). Here, we review glutamine metabolic pathways and how changes in cellular glutamine utilization enable the progression of RCC. This overview provides scientific rationale for targeting this pathway in patients with mRCC. We will summarize the current understanding of cellular and molecular mechanisms underlying anti-tumor efficacy of glutaminase inhibitors in RCC, provide an overview of clinical efforts targeting glutaminase in mRCC, and review approaches for identifying biomarkers for patient stratification and detecting therapeutic response early on in patients treated with this novel class of anti-cancer drug. Ultimately, results of ongoing clinical trials will demonstrate whether glutaminase inhibition can be a worthy addition to the current armamentarium of drugs used for patients with mRCC.
Collapse
Affiliation(s)
- Christian R Hoerner
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CA, USA
| | - Viola J Chen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CA, USA
| | - Alice C Fan
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CA, USA
| |
Collapse
|
8
|
Abstract
INTRODUCTION Four isomeric structures of thiadiazole motifs have outstanding pharmacological inhibitory applications are reported in this review. Thiadiazole nucleus is present in several biologically active natural products and commercial drugs. Most of thiadiazoles reported herein are emphasized to have broad spectrum of medicinal activities. Areas covered: This review represents the recent inhibitory activities of thiadiazole isomeric scaffolds and their broad-spectrum biological applications published as full texts during 2010-2016 as well as the patents published during 2005-2016. The inhibition areas covered included anti-inflammatory, antimicrobial, antitumor, antioxidant, antitubercular, antiviral, antileishmanial, anticonvulsant, herbicidal and algicidal activities in addition to enzymes, human platelet aggregation and neuroprotective inhibitors. Expert opinion: This survey revealed very interesting data about the applications of thiadiazoles, where some synthetic or natural thiadiazole derivatives were components of drugs available in the market. Many thiadiazole derivatives can be considered as lead compounds for drug synthesis. The most inhibitory active 1,3,4-thiadiazole compounds are those incorporating secondary alkyl(aryl)amido- and/or benzylthio(mercapto) groups at positions 2 and 5. Several thiadiazole derivatives demonstrated higher antibacterial, antitumor and antiviral activities than the standard drugs. Some thiadiazole derivatives exhibited high selective enzymes inhibitory activities based on the electronic properties of the substituents at positions 2 or 5.
Collapse
Affiliation(s)
- Kamal M Dawood
- a Department of Chemistry, Faculty of Science , Kuwait University , Safat , Kuwait.,b Department of Chemistry, Faculty of Science , Cairo University , Giza , Egypt
| | - Thoraya A Farghaly
- b Department of Chemistry, Faculty of Science , Cairo University , Giza , Egypt.,c Department of Chemistry, Faculty of Applied Science , Umm Al-Qura University , Makkah Almukkarramah , Saudi Arabia
| |
Collapse
|
9
|
van der Mijn JC, Panka DJ, Geissler AK, Verheul HM, Mier JW. Novel drugs that target the metabolic reprogramming in renal cell cancer. Cancer Metab 2016; 4:14. [PMID: 27418963 PMCID: PMC4944519 DOI: 10.1186/s40170-016-0154-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/30/2016] [Indexed: 02/07/2023] Open
Abstract
Molecular profiling studies of tumor tissue from patients with clear cell renal cell cancer (ccRCC) have revealed extensive metabolic reprogramming in this disease. Associations were found between metabolic reprogramming, histopathologic Fuhrman grade, and overall survival of patients. Large-scale genomics, proteomics, and metabolomic analyses have been performed to identify the molecular players in this process. Genes involved in glycolysis, the pentose phosphate pathway, glutamine metabolism, and lipogenesis were found to be upregulated in renal cell cancer (RCC) specimens as compared to normal tissue. Preclinical research indicates that mutations in VHL, FBP1, and the PI3K-AKT-mTOR pathway drives aerobic glycolysis through transcriptional activation of the hypoxia-inducible factors (HIF). Mechanistic studies revealed glutamine as an important source for de novo fatty acid synthesis through reductive carboxylation. Amplification of MYC drives reductive carboxylation. In this review, we present a detailed overview of the metabolic changes in RCC in conjunction with potential novel therapeutics. We discuss preclinical studies that have investigated targeted agents that interfere with various aspects of tumor cell metabolism and emphasize their impact specifically on glycolysis, lipogenesis, and tumor growth. Furthermore, we describe a number of phase 1 and 2 clinical trials that have been conducted with these agents.
Collapse
Affiliation(s)
- Johannes C van der Mijn
- Department of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215 USA ; Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands ; Department of Internal Medicine, OLVG; Jan van Tooropstraat 164, 1061 AE Amsterdam, The Netherlands
| | - David J Panka
- Department of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215 USA
| | - Andrew K Geissler
- Department of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215 USA
| | - Henk M Verheul
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - James W Mier
- Department of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA 02215 USA
| |
Collapse
|
10
|
Synthesis and antioxidant activity of a new class of sulfone/sulfonamide-linked bis(oxadiazoles), bis(thiadiazoles), and bis(triazoles). Med Chem Res 2014. [DOI: 10.1007/s00044-014-1277-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
11
|
Guda DR, Cho HM, Lee ME. Mild and convenient one-pot synthesis of 2-amino-1,3,4-thiadiazoles using trimethylsilyl isothiocyanate (TMSNCS). RSC Adv 2013. [DOI: 10.1039/c3ra00159h] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
12
|
A series of 2-arylamino-5-(indolyl)-1,3,4-thiadiazoles as potent cytotoxic agents. Eur J Med Chem 2012; 55:432-8. [DOI: 10.1016/j.ejmech.2012.06.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Revised: 06/07/2012] [Accepted: 06/22/2012] [Indexed: 11/20/2022]
|
13
|
Kumar D, Vaddula BR, Chang KH, Shah K. One-pot synthesis and anticancer studies of 2-arylamino-5-aryl-1,3,4-thiadiazoles. Bioorg Med Chem Lett 2011; 21:2320-3. [DOI: 10.1016/j.bmcl.2011.02.083] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Revised: 02/22/2011] [Accepted: 02/23/2011] [Indexed: 11/25/2022]
|
14
|
Efferth T. Research Highlights: Broken dreams or time to test? Chemoselective treatment of MTAP-deficient tumors with L-alanosine. Per Med 2009; 6:373-375. [DOI: 10.2217/pme.09.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Thomas Efferth
- German Cancer Research Center, Pharmaceutical Biology (C015), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Kindler HL, Burris HA, Sandler AB, Oliff IA. A phase II multicenter study of L-alanosine, a potent inhibitor of adenine biosynthesis, in patients with MTAP-deficient cancer. Invest New Drugs 2008; 27:75-81. [PMID: 18618081 DOI: 10.1007/s10637-008-9160-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Accepted: 06/24/2008] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Methylthioadenosine phosphorylase (MTAP)-deficient tumors are dependent on the de novo purine synthesis pathway. These cancers are potential targets for selective chemotherapy with inhibitors of de novo adenine synthesis such as L-alanosine [L-2-amino-3-(N-hydroxy-N-nitrosamino) propionic acid]. This phase II study was designed to evaluate the efficacy and safety of L-alanosine in patients with MTAP-deficient solid tumors. METHODS Patients with mesothelioma, non-small cell lung cancer (NSCLC), soft tissue sarcoma, osteosarcoma, or pancreatic cancer whose tumors were MTAP deficient by immunohistochemistry were eligible. Patients received L-alanosine at a starting dose of 80 mg/m(2) by continuous intravenous infusion daily for 5 days every 21 days. Computed tomography scans or magnetic resonance imaging were performed every 3 cycles. RESULTS 65 patients (16 mesothelioma, 13 NSCLC, 15 soft tissue sarcoma, 7 osteosarcoma, 14 pancreatic cancer) were enrolled at 19 centers; 55 were evaluable for response. There were no objective responses; 24% had s disease, including 2 patients with mesothelioma who had prolonged stable disease lasting 7.5 and 15.2 months, respectively. Grade 3/4 toxicities included mucositis 11%, fatigue 6%, nausea 3%, and renal failure 1.5%. CONCLUSION At this dose and schedule, L-alanosine was ineffective in patients with advanced MTAP-deficient tumors.
Collapse
Affiliation(s)
- Hedy Lee Kindler
- University of Chicago, 5841 South Maryland Avenue, MC2115, Chicago, IL, 60637-1460, USA.
| | | | | | | |
Collapse
|
16
|
Matysiak J, Opolski A. Synthesis and antiproliferative activity of N-substituted 2-amino-5-(2,4-dihydroxyphenyl)-1,3,4-thiadiazoles. Bioorg Med Chem 2006; 14:4483-9. [PMID: 16517170 DOI: 10.1016/j.bmc.2006.02.027] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2006] [Revised: 02/08/2006] [Accepted: 02/14/2006] [Indexed: 11/30/2022]
Abstract
A number of N-substituted 2-amino-5-(2,4-dihydroxyphenyl)-1,3,4-thiadiazoles were synthesized and evaluated for their antiproliferative activities. The panel substitution included alkyl, aryl, and morpholinoalkyl derivatives. The structures of compounds were identified from elemental, IR, (1)H NMR, (13)C NMR and MS spectra analyses. The cytotoxicity in vitro against the four human cell lines: SW707 (rectal), HCV29T (bladder), A549 (lung), and T47D (breast) was determined. Alkyl and morpholinoalkyl derivatives exhibited significantly lower effect than phenyl ones. The highest antiproliferative activity was found for 2-(2,4-dichlorophenylamino)-5-(2,4-dihydroxyphenyl)-1,3,4-thiadiazole, with ID(50) two times lower (SW707, T47D) than for cisplatin studied comparatively as the control compound.
Collapse
Affiliation(s)
- Joanna Matysiak
- Department of Chemistry, Agricultural University, Lublin, Poland.
| | | |
Collapse
|
17
|
Matysiak J, Nasulewicz A, Pełczyńska M, Switalska M, Jaroszewicz I, Opolski A. Synthesis and antiproliferative activity of some 5-substituted 2-(2,4-dihydroxyphenyl)-1,3,4-thiadiazoles. Eur J Med Chem 2006; 41:475-82. [PMID: 16517026 DOI: 10.1016/j.ejmech.2005.12.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Accepted: 12/08/2005] [Indexed: 10/25/2022]
Abstract
A series of new 5-substituted 2-(2,4-dihydroxyphenyl)-1,3,4-thiadiazoles has been synthesised and evaluated for their antiproliferative activity. The compounds were prepared by the reaction of the sulphinylbis(2,4-dihydroxythiobenzoyl) (STB) wit hydrazides or carbazates. The panel substitution included alkyl, alkoxy, aryl and heteroaryl derivatives. The structures of compounds were identified from the elemental, IR, (1)H NMR and MS spectra analysis. The highest antiproliferative activity against the cells of human cancer lines for 2-(2,4-dihydroxyphenyl)-5-(4-methoxybenzyloxy)-1,3,4-thiadiazole was found with ID(50) values comparable (HCV29T and SW707) or significantly lower (T47D) than for cisplatin applied as the reference compound. The influence of 5-substiution type of 2-(2,4-dihydroxyphenyl)-1,3,4-thiadiazoles on antiproliferative activity is discussed.
Collapse
Affiliation(s)
- Joanna Matysiak
- Department of Chemistry, Agricultural University, Lublin, Poland.
| | | | | | | | | | | |
Collapse
|
18
|
Karikari CA, Mullendore M, Eshleman JR, Argani P, Leoni LM, Chattopadhyay S, Hidalgo M, Maitra A. Homozygous deletions of methylthioadenosine phosphorylase in human biliary tract cancers. Mol Cancer Ther 2006; 4:1860-6. [PMID: 16373701 DOI: 10.1158/1535-7163.mct-05-0103] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The p16(INK4A)/CDKN2A gene on chromosome 9p21 is a site of frequent allelic loss in human cancers, and in a subset of cases, homozygous deletions at this locus encompass the telomeric methylthioadenosine phosphorylase (MTAP) gene. The MTAP gene product is the principal enzyme involved in purine synthesis via the salvage pathway, such that MTAP-negative cancers are solely dependent on de novo purine synthesis mechanisms. Inhibitors of the de novo pathway can then be used to selectively blockade purine synthesis in cancer cells while causing minimal collateral damage to normal cells. In this study, we determine that 10 of 28 (35%) biliary tract cancers show complete lack of Mtap protein expression. In vitro analysis using a selective inhibitor of the de novo purine synthesis pathway, L-alanosine, shows robust growth inhibition in MTAP-negative biliary cancer cell lines CAK-1 and GBD-1 accompanied by striking depletion of intracellular ATP and failure to rescue this depletion via addition of exogenous methylthioadenosine, the principal substrate of the MTAP gene product; in contrast, no significant effects were observed in MTAP-expressing HuCCT1 and SNU308 cell lines. Colony formation studies confirmed that L-alanosine reduced both number and size of CAK-1 colonies in soft agar assays. Knockdown of Mtap protein by RNA interference in L-alanosine-resistant HuCCT1 cells conferred sensitivity to this agent, confirming that intracellular Mtap protein levels determine response to L-alanosine. Inhibitors of de novo purine synthesis can be a potential mechanism-based strategy for treatment of biliary tract cancers, one third of which show complete loss of MTAP function.
Collapse
Affiliation(s)
- Collins A Karikari
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Li XM, Kanekal S, Crépin D, Guettier C, Carrière J, Elliott G, Lévi F. Circadian pharmacology of l-alanosine (SDX-102) in mice. Mol Cancer Ther 2006; 5:337-46. [PMID: 16505107 DOI: 10.1158/1535-7163.mct-05-0332] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
L-alanosine (SDX-102) exerts its cytotoxicity through inhibition of de novo purine biosynthesis, an effect potentiated by methylthioadenosine phosphorylase (MTAP) deficiency. The relevance of circadian dosing time was investigated for chronotherapeutic optimization of SDX-102. Toxicity was assessed in healthy mice following single (1,150, 1,650, or 1,850 mg/kg/d) or multiple doses (250 or 270 mg/kg/d). Efficacy was tested in mice with P388 leukemia receiving multiple doses (225 or 250 mg/kg/d). SDX-102 was administered at six circadian times 4 hours apart in mice synchronized with 12 hours of light alternating with 12 hours of darkness. MTAP expression was determined in liver, bone marrow, small intestinal mucosa, and P388 cells. Dosing at 19 hours after light onset reduced lethality 5-fold after single administration and 3-fold after multiple doses as compared with worst time [P < 0.001 and P < 0.01, respectively (chi2 test)]. Neutropenia, lymphopenia, and bone marrow hemorrhagic lesions were significantly less in mice dosed at 19 hours after light onset as compared with 7 hours after light onset. SDX-102 at 7 hours after light onset transiently ablated the 24-hour patterns in body temperature and activity. A circadian rhythm characterized small intestinal MTAP expression with a maximum at 6:30 hours after light onset (P = 0.04). A minor survival improvement was found in MTAP-deficient P388 mice receiving SDX-102 at 7 or 23 hours after light onset as compared with other times (P = 0.03, log-rank test). In conclusion, the therapeutic index of SDX-102 was improved by the delivery of SDX-102 in the mid to late activity span. These results support the concept of chronomodulated infusion of SDX-102 in cancer patients.
Collapse
Affiliation(s)
- Xiao-Mei Li
- Institut National de la Sante et de la Recherche Medicale, U776 "Rythmes biologiques et cancers" (Université Paris Sud), Paul Brousse Hospital, 94800 Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
20
|
Bates SE, Bakke S, Kang M, Robey RW, Zhai S, Thambi P, Chen CC, Patil S, Smith T, Steinberg SM, Merino M, Goldspiel B, Meadows B, Stein WD, Choyke P, Balis F, Figg WD, Fojo T. A phase I/II study of infusional vinblastine with the P-glycoprotein antagonist valspodar (PSC 833) in renal cell carcinoma. Clin Cancer Res 2005; 10:4724-33. [PMID: 15269145 DOI: 10.1158/1078-0432.ccr-0829-03] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE P-glycoprotein (Pgp) inhibitors have been under clinical evaluation for drug resistance reversal for over a decade. Valspodar (PSC 833) inhibits Pgp-mediated efflux but delays drug clearance, requiring reduction of anticancer drug dosage. We designed an infusional schedule for valspodar and vinblastine to mimic infusional vinblastine alone. The study was designed to determine the maximally tolerated dose of vinblastine, while attempting to understand the pharmacokinetic interactions between vinblastine and valspodar and to determine the response rate in patients with metastatic renal cell cancer. PATIENTS AND METHODS Thirty-nine patients received continuous infusion valspodar and vinblastine. Vinblastine was administered for 3 days to compensate for the expected delay in clearance and the required dose reduction. Valspodar was administered initially at a dose of 10 mg/kg/d; the dose of vinblastine varied. RESULTS The maximum-tolerated dose of vinblastine was 1.3 mg/m(2)/d. As suggested previously, serum valspodar concentrations exceeded those needed for Pgp inhibition. Consequently, the dose of valspodar was reduced to 5 mg/kg, allowing a vinblastine dose of 2.1 mg/m(2)/d to be administered. Pharmacodynamic studies demonstrated continued inhibition of Pgp at lower valspodar doses by functional assay in Pgp-expressing CD56+ cells and by (99m)Tc-sestamibi imaging. A 15-fold range in cytochrome p450 activity was observed, as measured by midazolam clearance. No major responses were observed. CONCLUSIONS These results suggest that the pharmacokinetic impact of cytochrome P450 inhibition by valspodar can be reduced although not eliminated, while preserving Pgp inhibition, thus separating the pharmacokinetic and pharmacodynamic activities of valspodar.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- Adolescent
- Adult
- Aged
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/adverse effects
- Antineoplastic Agents, Phytogenic/pharmacokinetics
- Area Under Curve
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Constipation/chemically induced
- Cyclosporins/administration & dosage
- Cyclosporins/adverse effects
- Cyclosporins/pharmacokinetics
- Dose-Response Relationship, Drug
- Fatigue/chemically induced
- Female
- Humans
- Infusions, Intravenous
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Male
- Metabolic Clearance Rate
- Middle Aged
- Treatment Outcome
- Vinblastine/administration & dosage
- Vinblastine/adverse effects
- Vinblastine/pharmacokinetics
Collapse
Affiliation(s)
- Susan E Bates
- Cancer Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Traditional cytotoxic chemotherapy has been considered to be ineffective in renal cell carcinoma, likely due to multiple mechanisms of high-level drug resistance proteins such as p-glycoprotein expressed by these cancers. Nonetheless, low level activity of several nucleoside analogues and the elucidation of critical molecular pathways and targets in this disease, such as the angiogenic pathway, provide hope that important advances can and will be made.
Collapse
Affiliation(s)
- Christopher M George
- Section of Hematology/Oncology, University of Chicago Medical Center, 5841 South Maryland Ave, MC 2115, Chicago, IL 60637-1470, USA.
| | | |
Collapse
|
22
|
Bates S, Kang M, Meadows B, Bakke S, Choyke P, Merino M, Goldspiel B, Chico I, Smith T, Chen C, Robey R, Bergan R, Figg WD, Fojo T. A Phase I study of infusional vinblastine in combination with the P-glycoprotein antagonist PSC 833 (valspodar). Cancer 2001; 92:1577-90. [PMID: 11745237 DOI: 10.1002/1097-0142(20010915)92:6<1577::aid-cncr1484>3.0.co;2-h] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND PSC 833 is a second-generation P-glycoprotein (Pgp) antagonist developed to reverse multidrug resistance (MDR). The authors conducted a Phase I study of orally administered PSC 833 in combination with vinblastine administered as a 5-day continuous infusion. METHODS Seventy-nine patients with advanced malignant disease were enrolled in the trial and treated with escalating doses of PSC 833. Pharmacokinetic interactions between PSC 833 and vinblastine were anticipated. Accordingly, when dose limiting toxicities were observed, the dose of vinblastine was reduced as PSC 833 was escalated. Three schedules and two formulations of PSC 833 were used in the study. RESULTS The maximum tolerated doses of PSC 833 were 12.5 mg/kg orally every 12 hours for 8 days for the liquid formulation in combination with 0.9 mg/m(2) per day vinblastine as a continuous intravenous infusion (CIV) for 5 days; and 4 mg/kg orally every 6 hours for 8 days for the microemulsion formulation in combination with 0.6 mg/m(2) per day vinblastine CIV for 5 days. The principal toxicities for PSC 833 were ataxia and paresthesias and for the combination, constipation, fever. and neutropenia. Increased oral bioavailability and increased peak and trough concentrations were observed with the microemulsion formulation. Significant interpatient variability in pharmacokinetic parameters was observed. Ten patients studied at the MTD for PSC 833 (4 mg/kg orally every 6 hours for 8 days) had inhibition of rhodamine efflux from CD56 positive peripheral lymphocytes as a surrogate for Pgp antagonism. Among 43 evaluable patients with clear cell carcinoma of the kidney, 3 patients had complete responses, and 1 patient had a partial response. CONCLUSIONS PSC 833 in combination with vinblastine can be administered safely to patients provided the vinblastine dose is adjusted for pharmacokinetic interactions. The high interpatient variability is a significant confounding factor. Surrogate studies with CD56 positive cells suggest that Pgp inhibition in the clinical setting is achievable. Improved methods for predicting pharmacokinetic interactions should improve future studies.
Collapse
Affiliation(s)
- S Bates
- Cancer Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Aberkane H, Frank P, Galteau MM, Wellman M. Acivicin induces apoptosis independently of gamma-glutamyltranspeptidase activity. Biochem Biophys Res Commun 2001; 285:1162-7. [PMID: 11478776 DOI: 10.1006/bbrc.2001.5297] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inhibition of cellular gamma-glutamyltranspeptidase (GGT) enzyme activity by its specific inhibitor acivicin is frequently used in studies aimed at demonstrating the physiological role of this enzyme. However, because acivicin is a glutamine antagonist, it also inhibits many other glutamine-dependent enzymes involved in purine and pyrimidine biosynthesis. The objective of the present work is to determine whether acivicin exhibits apoptotic properties and the significance of GGT activity level in the response to acivicin treatment. We compared acivicin (0-150 microM) effect on V79 cell lines expressing or not expressing human GGT. Apoptosis was assayed by annexin-V staining, cell cycle analysis, and caspase activation using flow cytometry. We found that acivicin causes a dose- and time-dependent apoptosis in the GGT-negative V79 cell line as well as in its GGT-positive counterpart line. This is the evidence that acivicin induces apoptosis in V79 cell independently of their GGT activity level.
Collapse
Affiliation(s)
- H Aberkane
- Thiols et Fonctions Cellulaires, Faculty of Pharmacy, Laboratoire de Biochimie, 30, rue Lionnois, Nancy, 54000, France
| | | | | | | |
Collapse
|
24
|
Abstract
Renal cell carcinoma (RCC) is characterized by (a) lack of early warning signs, which results in a high proportion of patients with metastases at the time of diagnosis; (b) protean clinical manifestations; and (c) resistance to radiotherapy and chemotherapy. The estimates of new diagnoses and deaths from kidney cancer in the United States during 1996 are 30,600 and 12,000, respectively. RCC occurs nearly twice as often in men as in women. The age at diagnosis is generally older than 40 years; the median age is in the midsixties. The incidence of RCC has been rising steadily. Between 1974 and 1990, there was a 38% increase in the number of patients who had a diagnosis of RCC. This increase was accompanied by a significant improvement in 5-year survival. Both trends are likely the result of improved diagnostic capability. Newer radiographic techniques, including ultrasonography, computed tomography, and magnetic resonance imaging, are detecting kidney tumors more frequently and at a lower disease stage, when tumors can be resected for cure. Surgical treatment is the only curative therapy for localized RCC. Radical nephrectomy remains the mainstay of surgical management, but techniques are being modified. These modifications include partial nephrectomy and resection of vena caval thrombi. In highly selected cases, surgical resection of locally recurrent RCC or of disease at a solitary metastatic site is associated with long-term survival. Metastatic RCC is highly resistant to the many systemic therapies that have been extensively investigated. A minority of patients achieve complete or partial response to interferon, interleukin-2, or both. Response can be dramatic but is rarely durable. Because most patients do not achieve response, these agents are not considered effective treatments for RCC, but the response in some patients indicates the need for continued research on their use. Identification of new agents with better antitumor activity against metastases remains a high priority in clinical investigation of therapy for this refractory disease.
Collapse
Affiliation(s)
- R J Motzer
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | |
Collapse
|
25
|
Witte RS, Elson P, Stewart JA, Malliard JA, Oken MM, Loehrer PJ. A phase II trial of melphalan or thioguanine in the treatment of patients with advanced renal cell cancer. Urol Oncol 1996; 2:96-8. [DOI: 10.1016/s1078-1439(96)00073-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
26
|
Prognostic Factors for Survival in Patients with Metastatic Renal Cancer Treated with Biological Response Modifiers. J Urol 1995. [DOI: 10.1097/00005392-199507000-00014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Prognostic Factors for Survival in Patients with Metastatic Renal Cancer Treated with Biological Response Modifiers. J Urol 1995. [DOI: 10.1016/s0022-5347(01)67218-5] [Citation(s) in RCA: 109] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
|
29
|
Yagoda A, Petrylak D, Thompson S. CYTOTOXIC CHEMOTHERAPY FOR ADVANCED RENAL CELL CARCINOMA. Urol Clin North Am 1993. [DOI: 10.1016/s0094-0143(21)00489-4] [Citation(s) in RCA: 138] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
30
|
Witte RS, Elson P, Bryan GT, Trump DL. Trimetrexate in advanced renal cell carcinoma. An ECOG phase II trial. Invest New Drugs 1992; 10:51-4. [PMID: 1535065 DOI: 10.1007/bf01275483] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Thirty-four chemotherapy-naive, ambulatory patients with advanced renal cell cancer were treated with the non-classical antifol trimetrexate at the intravenous dose of 12 mg/m2 daily x 5 every three weeks (8 mg/m2 qd x 5 for greater than 30% bone marrow previously irradiated). One patient experienced a partial response lasting 24 weeks for a response rate of 3% (exact 95% CI, 0.1 to 15.3%). Toxicity was manageable and primarily myelosuppression, gastrointestinal, and mucosal. Trimetrexate has little activity in advanced renal cell carcinoma at this dose and schedule.
Collapse
Affiliation(s)
- R S Witte
- University of Wisconsin Clinical Cancer Center, Madison
| | | | | | | |
Collapse
|
31
|
Rowinsky EK, Donehower RC. The clinical pharmacology and use of antimicrotubule agents in cancer chemotherapeutics. Pharmacol Ther 1991; 52:35-84. [PMID: 1687171 DOI: 10.1016/0163-7258(91)90086-2] [Citation(s) in RCA: 220] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Although there has been a rapid expansion of the number of classes of compounds with antineoplastic activity, few have played a more vital role in the curative and palliative treatment of cancers than the antimicrotubule agents. Although the vinca alkaloids have been the only subclass of antimicrotubule agents that have had broad experimental and clinical applications in oncologic therapeutics over the last several decades, the taxanes, led by the prototypic agent taxol, are emerging as another very active class of antimicrotubule agents. After briefly reviewing the mechanisms of antineoplastic action and resistance, this article comprehensively reviews the clinical pharmacology, therapeutic applications, and clinical toxicities of selected antimicrotubule agents.
Collapse
Affiliation(s)
- E K Rowinsky
- Division of Pharmacology and Experimental Therapeutics, Johns Hopkins Oncology Center, Baltimore, Maryland 21205
| | | |
Collapse
|
32
|
Heicappell R, Ackermann R. Rationale for immunotherapy of renal cell carcinoma. UROLOGICAL RESEARCH 1990; 18:357-72. [PMID: 2100410 DOI: 10.1007/bf00297367] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Metastasis to distant organs is the principal cause of death from renal cell carcinoma (RCC). No commonly accepted therapy is available for disseminated RCC at present. Immunotherapy is a mode of therapy that either interferes with the immune system or makes use of drugs that have been derived from soluble mediators of the immune system. Several lines of evidence suggest that combinations of genetically engineered cytokines (e.g. interleukin-2 and interferon alpha) may be particularly active in the treatment of advanced RCC. There are two major rationales for considering immunotherapy for RCC: (1) there is currently no other therapy available, and (2) there is hardly any innovative approach besides immunotherapy. Still, immunotherapy is far from being a standard therapy for disseminated RCC.
Collapse
Affiliation(s)
- R Heicappell
- Department of Urology, Heinrich-Heine-University, Düsseldorf, FRG
| | | |
Collapse
|