1
|
de la Torre AN, Contractor S, Castaneda I, Cathcart CS, Razdan D, Klyde D, Kisza P, Gonzales SF, Salazar AM. A Phase I trial using local regional treatment, nonlethal irradiation, intratumoral and systemic polyinosinic-polycytidylic acid polylysine carboxymethylcellulose to treat liver cancer: in search of the abscopal effect. J Hepatocell Carcinoma 2017; 4:111-121. [PMID: 28848723 PMCID: PMC5557908 DOI: 10.2147/jhc.s136652] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose To determine the safety of an approach to immunologically enhance local treatment of hepatocellular cancer (HCC) by combining nonlethal radiation, local regional therapy with intratumoral injection, and systemic administration of a potent Toll-like receptor (TLR) immune adjuvant. Methods Patients with HCC not eligible for liver transplant or surgery were subject to: 1) 3 fractions of 2-Gy focal nonlethal radiation to increase tumor antigen expression, 2) intra-/peri-tumoral (IT) injection of the TLR3 agonist, polyinosinic-polycytidylic acid polylysine carboxymethylcellulose (poly-ICLC), to induce an immunologic “danger” response in the tumor microenvironment with local regional therapy, and 3) systemic boosting of immunity with intramuscular poly-ICLC. Primary end points were safety and tolerability; secondary end points were progression-free survival (PFS) and overall survival (OS) at 6 months, 1 year, and 2 years. Results Eighteen patients with HCC not eligible for surgery or liver transplant were treated. Aside from 1 embolization-related severe adverse event, all events were ≤grade II. PFS was 66% at 6 months, 39% at 12 months, and 28% at 24 months. Overall 1-year survival was 69%, and 2-year survival 38%. In patients <60 years old, 2-year survival was 62.5% vs. 11.1% in patients aged >60 years (P<0.05). Several patients had prolonged PFS and OS. Conclusion Intra-tumoral injection of the TLR3 agonist poly-ICLC in patients with HCC is safe and tolerable when combined with local nonlethal radiation and local regional treatment. Further work is in progress to evaluate if this approach improves survival compared to local regional treatment alone and characterize changes in anticancer immunity.
Collapse
Affiliation(s)
- Andrew N de la Torre
- Department of Surgery, St Joseph's Regional Medical Center, Paterson.,Department of Surgery, Rutgers New Jersey Medical School-University Hospital
| | - Sohail Contractor
- Department of Interventional Radiology, Rutgers New Jersey Medical School-University Hospital
| | - Ismael Castaneda
- Department of Surgery, St Joseph's Regional Medical Center, Paterson
| | | | - Dolly Razdan
- Department of Radiation Oncology, Clara Maas Hospital, Belleville, NJ
| | - David Klyde
- Department of Interventional Radiology, Rutgers New Jersey Medical School-University Hospital
| | - Piotr Kisza
- Department of Interventional Radiology, Rutgers New Jersey Medical School-University Hospital
| | - Sharon F Gonzales
- Department of Interventional Radiology, Rutgers New Jersey Medical School-University Hospital
| | | |
Collapse
|
2
|
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
|
3
|
Allard CB, Gelpi-Hammerschmidt F, Harshman LC, Choueiri TK, Faiena I, Modi P, Chung BI, Tinay I, Singer EA, Chang SL. Contemporary trends in high-dose interleukin-2 use for metastatic renal cell carcinoma in the United States. Urol Oncol 2015. [PMID: 26210683 DOI: 10.1016/j.urolonc.2015.06.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Targeted therapies (TTs) have revolutionized metastatic renal cell carcinoma (mRCC) treatment in the past decade, largely replacing immunotherapy including high-dose interleukin-2 (HD IL-2) therapy. We evaluated trends in HD IL-2 use for mRCC in the TT era. METHODS Our cohort comprised a weighted estimate of all patients undergoing HD IL-2 treatment for mRCC from 2004 to 2012 using the Premier Hospital Database. We assessed temporal trends in HD IL-2 use including patient, disease, and hospital characteristics stratified by era (pre-TT uptake: 2004-2006, uptake: 2007-2009, and post-TT uptake: 2010-2012) and fitted multivariable regression models to identify predictors of treatment toxicity and tolerability. RESULTS An estimated 2,351 patients received HD IL-2 therapy for mRCC in the United States from 2004 to 2012. The use decreased from 2004 to 2008. HD IL-2 therapy became increasingly centralized in teaching hospitals (24% of treatments in 2004 and 89.5% in 2012). Most patients who received HD IL-2 therapy were men, white, younger than 60 years, had lung metastases, and were otherwise healthy. Vasopressors, intensive care unit admission, and hemodialysis were necessary in 53.4%, 33.0%, and 7.1%, respectively. Factors associated with toxicities in multivariable analyses included being unmarried, male sex, and multiple metastatic sites. African Americans and patients with single-site metastases were less likely to receive multiple treatment cycles. CONCLUSIONS HD IL-2 therapy is used infrequently for mRCC in the United States, and its application has diminished with the uptake of TT. Patients are being increasingly treated in teaching hospitals, suggesting a centralization of care and possible barriers to access. A recent slight increase in HD IL-2 therapy use likely reflects recognition of the inability of TT to effect a complete response.
Collapse
Affiliation(s)
- Christopher B Allard
- Division of Urology, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA; Department of Urology, Massachusetts General Hospital, Boston, MA.
| | - Francisco Gelpi-Hammerschmidt
- Division of Urology, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA; Department of Urology, Massachusetts General Hospital, Boston, MA
| | - Lauren C Harshman
- Kidney Cancer Center, Dana-Farber Cancer Institute, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA
| | - Toni K Choueiri
- Kidney Cancer Center, Dana-Farber Cancer Institute, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA
| | - Izak Faiena
- Section of Urologic Oncology, Rutgers Institute of New Jersey and Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Parth Modi
- Section of Urologic Oncology, Rutgers Institute of New Jersey and Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Benjamin I Chung
- Department of Urology, Stanford School of Medicine, Palo Alto, CA
| | - Ilker Tinay
- Division of Urology, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA; Department of Urology, Marmara University School of Medicine, Istanbul, Turkey
| | - Eric A Singer
- Section of Urologic Oncology, Rutgers Institute of New Jersey and Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Steven L Chang
- Division of Urology, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
4
|
Figlin RA. Personalized immunotherapy (AGS-003) when combined with sunitinib for the treatment of metastatic renal cell carcinoma. Expert Opin Biol Ther 2015; 15:1241-8. [DOI: 10.1517/14712598.2015.1063610] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
5
|
Survival with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): Phase 2 study results. J Immunother Cancer 2015; 3:14. [PMID: 25901286 PMCID: PMC4404644 DOI: 10.1186/s40425-015-0055-3] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 03/02/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AGS-003 is an autologous immunotherapy prepared from fully matured and optimized monocyte-derived dendritic cells, which are co-electroporated with amplified tumor RNA plus synthetic CD40L RNA. AGS-003 was evaluated in combination with sunitinib in an open label phase 2 study in intermediate and poor risk, treatment naïve patients with metastatic clear cell renal cell carcinoma (mRCC). METHODS Twenty-one intermediate and poor risk patients were treated continuously with sunitinib (4 weeks on, 2 weeks off per 6 week cycle). After completion of the first cycle of sunitinib, patients were treated with AGS-003 every 3 weeks for 5 doses, then every 12 weeks until progression or end of study. The primary endpoint was to determine the complete response rate. Secondary endpoints included clinical benefit, safety, progression free survival (PFS) and overall survival (OS). Immunologic response was also monitored. RESULTS Thirteen patients (62%) experienced clinical benefit (9 partial responses, 4 with stable disease); however there were no complete responses in this group of intermediate and poor risk mRCC patients and enrollment was terminated early. Median PFS from registration was 11.2 months (95% CI 6.0, 19.4) and the median OS from registration was 30.2 months (95% CI 9.4, 57.1) for all patients. Seven (33%) patients survived for at least 4.5 years, while five (24%) survived for more than 5 years, including 2 patients who remain progression-free with durable responses for more than 5 years at the time of this report. AGS-003 was well tolerated with only mild injection-site reactions. The most common adverse events were related to expected toxicity from sunitinib therapy. In patients who had sequential samples available for immune monitoring, the magnitude of the increase in the absolute number of CD8(+) CD28(+) CD45RA(-) effector/memory T cells (CTLs) after 5 doses of AGS-003 relative to baseline, correlated with overall survival. CONCLUSIONS AGS-003 in combination with sunitinib was well tolerated and yielded supportive immunologic responses coupled with extension of median and long-term survival in an unselected, intermediate and poor risk prognosis mRCC population. CLINICAL TRIAL REGISTRY #NCT00678119.
Collapse
|
6
|
Dharmasiri U, Isenberg SL, Glish GL, Armistead PM. Differential ion mobility spectrometry coupled to tandem mass spectrometry enables targeted leukemia antigen detection. J Proteome Res 2014; 13:4356-62. [PMID: 25184817 PMCID: PMC4184456 DOI: 10.1021/pr500527c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Differential ion mobility spectrometry (DIMS) can be used as a filter to remove undesired background ions from reaching the mass spectrometer. The ability to use DIMS as a filter for known analytes makes DIMS coupled to tandem mass spectrometry (DIMS-MS/MS) a promising technique for the detection of cancer antigens that can be predicted by computational algorithms. In experiments using DIMS-MS/MS that were performed without the use of high-performance liquid chromatography (HPLC), a predicted model antigen, GLR (FLSSANEHL), was detected at a concentration of 10 pM (20 amol) in a mixture containing 94 competing model peptide antigens, each at a concentration of 1 μM. Without DIMS filtering, the GLR peptide was undetectable in the mixture even at 100 nM. Again, without using HPLC, DIMS-MS/MS was used to detect 2 of 3 previously characterized antigens produced by the leukemia cell line U937.A2. Because of its sensitivity, a targeted DIMS-MS/MS methodology can likely be used to probe for predicted cancer antigens from cancer cell lines as well as human tumor samples.
Collapse
Affiliation(s)
- Udara Dharmasiri
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill , 450 West Drive, 21-244, Chapel Hill, North Carolina 27599, United States
| | | | | | | |
Collapse
|
7
|
Hanzly M, Aboumohamed A, Yarlagadda N, Creighton T, Digiorgio L, Fredrick A, Rao G, Mehedint D, George S, Attwood K, Kauffman E, Narashima D, Khushalani NI, Pili R, Schwaab T. High-dose interleukin-2 therapy for metastatic renal cell carcinoma: a contemporary experience. Urology 2014; 83:1129-34. [PMID: 24767525 DOI: 10.1016/j.urology.2014.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 01/28/2014] [Accepted: 02/03/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To present our experience of high-dose interleukin-2 (HDIL-2) in a high-volume National Cancer Institute-designated center for patients with metastatic renal cell carcinoma (mRCC). METHODS Patients with mRCC who received HDIL-2 monotherapy as a first- or second-line therapy during 2004-2011 were identified. Demographics, pathologic variables, renal function, time until the start of HDIL-2 therapy, number of cycles (1-3), responses (complete response, partial response, stable disease, and progressive disease), and primary renal cell carcinoma treatment were analyzed. Progression-free survival and overall survival (OS) were determined. RESULTS Of 906 patients in the kidney cancer database, 91 patients with mRCC were treated with HDIL-2 and 18 patients (20.5%) underwent prior cytoreductive nephrectomy. Median age was 51 years, and 73.9% were men. Median follow-up was 45 months. Pretreatment renal function impairment led to more treatment cycles (2-3) than in those with adequate initial kidney function (92.3% vs 50.6%, respectively; P = .002). Lower tumor stage correlated with a better response (P = .023) and with longer time from diagnosis to initiation of HDIL-2 (P = .011). Complications included hypotension (67.4%), renal impairment (63%), impaired liver function (42.4%), and thrombocytopenia (31.5%). Four patients (4.5%) had a complete response, 10 (11.4%) had a partial response, and 28 (31.8%) had a stable disease. Median progression-free survival and OS were 8.6 and 35.5 months, respectively. The estimated 2-year OS rate was 60.6%. CONCLUSION Incorporating HDIL-2 therapy in the treatment strategies for mRCC added to the patients' survival in this series. HDIL-2 therapy is well tolerated in patients with pre-existing renal impairment with no long-term renal toxicity.
Collapse
Affiliation(s)
- Michael Hanzly
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY.
| | | | | | | | | | - Ariel Fredrick
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY
| | - Gaurav Rao
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY
| | - Diana Mehedint
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY
| | - Saby George
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY
| | | | - Eric Kauffman
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY
| | | | | | - Roberto Pili
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY
| | - Thomas Schwaab
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY
| |
Collapse
|
8
|
Bernstein MB, Garnett CT, Zhang H, Velcich A, Wattenberg MM, Gameiro SR, Kalnicki S, Hodge JW, Guha C. Radiation-induced modulation of costimulatory and coinhibitory T-cell signaling molecules on human prostate carcinoma cells promotes productive antitumor immune interactions. Cancer Biother Radiopharm 2014; 29:153-61. [PMID: 24693958 DOI: 10.1089/cbr.2013.1578] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We sought to determine if single-dose external beam radiation therapy (EBRT) could modulate the expression signature of T-cell costimulatory and coinhibitory molecules in human prostate cancer (PCa) cell lines in vitro. We investigated the functional impact of irradiated PCa cells with a modulated costimulatory profile on responder T-cell activity. We used three PCa cell lines (DU145, PC3, and LNCaP) and two epithelial cell lines from noncancerous prostate and lung tissue. After 72 hours of EBRT, surface expression of four immunostimulatory molecules (CD70, CD275/ICOSL, CD134L/OX40L, and CD137L/41BBL) and two immunosuppressive markers (CTLA-4/CD152 and PD-L1/CD274) were evaluated by flow cytometry. We evaluated the impact of several radiation doses and the longevity of modulated expression. We examined the functional impact of radiation-induced modulation of cancer cells by cytotoxic T cells (CTL) cytotoxicity and ELISPOT assay for interferon-gamma (IFN-γ) production. Last, we evaluated whether IFN-γ-induced PD-L1 expression could be reversed by EBRT. After 10 Gy EBRT, expression of OX40L and 41BBL increased in all three PCa cell lines; expression of CD70 and ICOSL increased in PC3 cells. Conversely, a decrease in PD-L1 expression in DU145 and PC3 cells was detectable up to 144 hours after EBRT. No PD-L1 was detected in LNCaP. Epithelial cells from normal prostate were not modulated by radiation. CTL cytolytic activity and IFN-γ production were enhanced by interaction with irradiated PCa cells. Finally, EBRT failed to prevent IFN-γ-induced upregulation of PD-L1. We demonstrate that a single dose of EBRT increased surface expression of costimulatory molecules and decreased the expression of coinhibitory molecules in human PCa cell lines. Changes in irradiated tumor cells led to functional enhancement of T-cell activity, despite EBRT failing to reduce IFN-γ-induced expression of PD-L1. These data suggest that combining radiotherapy with T-cell stimulating immunotherapy may be an attractive strategy for cancer treatment.
Collapse
Affiliation(s)
- Michael B Bernstein
- 1 Department of Radiation Oncology, Montefiore Medical Center , Albert Einstein College of Medicine, Bronx, New York
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Figlin RA. A novel personalized vaccine approach in combination with targeted therapy in advanced renal cell carcinoma. Immunotherapy 2013; 6:261-8. [PMID: 24354908 DOI: 10.2217/imt.13.168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The historical treatment paradigm for metastatic renal cell carcinoma has focused on immunomodulatory agents, such as IFN-α and IL-2, which provide good clinical outcomes in only a subset of patients. The development of therapies that target the VEGF and mTOR pathways have significantly altered the treatment landscape for this disease, with novel inhibitors providing substantial improvements in progression-free and overall survival over previous standards of care. Despite these advances, toxicity from targeted therapy and the development of resistance results in disease progression. By contrast, vaccine-based immunotherapy represents a promising new approach for the treatment of patients with metastatic renal cell carcinoma; however, tumor-induced immunosuppression has limited the clinical efficacy of this modality until recently. Some evidence suggests that certain targeted therapies, such as sunitinib, may reduce this immunosuppression and enhance the tumor microenvironment to promote synergy with autologous dendritic cell vaccines.
Collapse
Affiliation(s)
- Robert A Figlin
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Saperstein Critical Care Tower 1S28, Los Angeles, CA 90048, USA.
| |
Collapse
|
10
|
Gunawardane ND, Vaghani SP, Kuzel TM, Cotliar JA. Acute generalized exanthematous pustulosis in a patient receiving high-dose recombinant interleukin-2. J Am Acad Dermatol 2013; 69:e183-4. [PMID: 24034391 DOI: 10.1016/j.jaad.2013.04.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 03/30/2013] [Accepted: 04/02/2013] [Indexed: 10/26/2022]
Affiliation(s)
- Nilanthi D Gunawardane
- Department of Dermatology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | | |
Collapse
|
11
|
Sittig SP, Køllgaard T, Grønbæk K, Idorn M, Hennenlotter J, Stenzl A, Gouttefangeas C, Thor Straten P. Clonal expansion of renal cell carcinoma-infiltrating T lymphocytes. Oncoimmunology 2013; 2:e26014. [PMID: 24228230 PMCID: PMC3820815 DOI: 10.4161/onci.26014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/02/2013] [Indexed: 11/19/2022] Open
Abstract
T lymphocytes can mediate the destruction of cancer cells by virtue of their ability to recognize tumor-derived antigenic peptides that are presented on the cell surface in complex with HLA molecules and expand. Thus, the presence of clonally expanded T cells within neoplastic lesions is an indication of ongoing HLA-restricted T cell-mediated immune responses. Multiple tumors, including renal cell carcinomas (RCCs), are often infiltrated by significant amounts of T cells, the so-called tumor-infiltrating lymphocytes (TILs). In the present study, we analyzed RCC lesions (n = 13) for the presence of expanded T-cell clonotypes using T-cell receptor clonotype mapping. Surprisingly, we found that RCCs comprise relatively low numbers of distinct expanded T-cell clonotypes as compared with melanoma lesions. The numbers of different T-cell clonotypes detected among RCC-infiltrating lymphocytes were in the range of 1–17 (median = 5), and in several patients, the number of clonotypes expanded within tumor lesions resembled that observed among autologous peripheral blood mononuclear cells. Moreover, several of these clonotypes were identical in TILs and PBMCs. Flow cytometry data demonstrated that the general differentiation status of CD8+ TILs differed from that of circulating CD8+ T cells. Furthermore, PD-1 and LAG-3 were expressed by a significantly higher percentage of CD8+ RCC-infiltrating lymphocytes as compared with PBMCs obtained from RCC patients or healthy individuals. Thus, CD8+ TILs display a differentiated phenotype and express activation markers as well as surface molecules associated with the inhibition of T-cell functions. However, TILs are characterized by a low amount of expanded T-cell clonotypes.
Collapse
Affiliation(s)
- Simone P Sittig
- Center for Cancer Immune Therapy (CCIT); Department of Hematology; Copenhagen University Hospital Herlev; Herlev, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Hsp90-binding immunophilins as a potential new platform for drug treatment. Future Med Chem 2013; 5:591-607. [PMID: 23573975 DOI: 10.4155/fmc.13.7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Immunophilins are proteins that contain a PPIase domain as a family signature. Low-molecular-weight immunophilins were first described associated to immunosuppressive action and protein folding. Recent studies of other members of the family have led to the identification of their participation in basic processes such as protein-protein interactions, signal transduction cascades, cell differentiation, cell cycle progression, metabolic activity, apoptosis mechanisms, microorganisms infection, cancer, neurotrophism and neuroprotection, among several other physiological and pathophysiological processes. Due to all these emerging features, the development of specific ligands for immunophilins appears to have promising perspectives, in particular in the fields of cancer biology and neuroregeneration fields. We review the emerging role of immunophilins in protein transport, transcription regulation, malignancies development and neurotrophic action, in addition to a number of biological properties that transform these proteins in potential targets for novel therapeutic strategies.
Collapse
|
13
|
Meyer A, Gruber A, Klopfleisch R. All Subunits of the Interleukin-2 Receptor are Expressed by Canine Cutaneous Mast Cell Tumours. J Comp Pathol 2013; 149:19-29. [DOI: 10.1016/j.jcpa.2012.11.232] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 09/25/2012] [Accepted: 11/12/2012] [Indexed: 11/26/2022]
|
14
|
Clark JM, Kelley B, Titze J, Fung H, Maciejewski J, Nathan S, Rich E, Basu S, Kaufman HL. Clinical and Safety Profile of High-Dose Interleukin-2 Treatment in Elderly Patients with Metastatic Melanoma and Renal Cell Carcinoma. Oncology 2013; 84:123-6. [DOI: 10.1159/000342764] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 07/05/2012] [Indexed: 11/19/2022]
|
15
|
Words of wisdom. Re: Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Eur Urol 2012; 62:182-3. [PMID: 22640859 DOI: 10.1016/j.eururo.2012.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
16
|
Giuliani J, Drudi F. Immunotherapy and targeted therapies in metastatic renal cell carcinoma: is there a preferred sequence? Cancer Biother Radiopharm 2012; 27:513-8. [PMID: 22738404 DOI: 10.1089/cbr.2012.1250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
INTRODUCTION Currently, the best sequence of targeted therapy in patients with metastatic renal cell carcinoma (mRCC) has not been sufficiently defined and is based on the patient's and physician's decision, which may be influenced by comorbidities and toxicity profiles. The aim of this study was to evaluate the outcome of target therapies on clinical practice after the era of cytokine-based therapy in mRCC. MATERIALS AND METHODS We retrospectively analyzed all consecutive patients with mRCC treated at our Clinical Oncology Unit from June 1998 to September 2010. RESULTS We evaluated 61 patients: 21 (34.4%) with only cytokine-based therapy (95.2% interferon-α), 24 (39.3%) with target therapies in first line (100% sunitinib), and 16 (26.2%) with target therapies in second or subsequent line. Median time follow-up was 16.18 months (range 2.1-171.1). Considering the type of therapy, the univariate analysis for overall survival showed statistically significant advantages for the use of target therapies in second or subsequent line (p=0.024). CONCLUSIONS Our data and consequently our proposal to revaluate the role of immunotherapy (also with the possibility of adding bevacizumab) in the first line are heavily provocative to point out the attention to this actually partially unsolved question; other larger experiences, pre-eminent opinion, and clinical trials are needed.
Collapse
Affiliation(s)
- Jacopo Giuliani
- Clinical Oncology Unit, St. Anna University Hospital , Ferrara, Italy.
| | | |
Collapse
|
17
|
Liang X, De Vera ME, Buchser WJ, Romo de Vivar Chavez A, Loughran P, Stolz DB, Basse P, Wang T, Van Houten B, Zeh HJ, Lotze MT. Inhibiting systemic autophagy during interleukin 2 immunotherapy promotes long-term tumor regression. Cancer Res 2012; 72:2791-801. [PMID: 22472122 PMCID: PMC3417121 DOI: 10.1158/0008-5472.can-12-0320] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Administration of high-dose interleukin-2 (HDIL-2) has durable antitumor effects in 5% to 10% of patients with melanoma and renal cell carcinoma. However, treatment is often limited by side effects, including reversible, multiorgan dysfunction characterized by a cytokine-induced systemic autophagic syndrome. Here, we hypothesized that the autophagy inhibitor chloroquine would enhance IL-2 immunotherapeutic efficacy and limit toxicity. In an advanced murine metastatic liver tumor model, IL-2 inhibited tumor growth in a dose-dependent fashion. These antitumor effects were significantly enhanced upon addition of chloroquine. The combination of IL-2 with chloroquine increased long-term survival, decreased toxicity associated with vascular leakage, and enhanced immune cell proliferation and infiltration in the liver and spleen. HDIL-2 alone increased serum levels of HMGB1, IFN-γ, IL-6, and IL-18 and also induced autophagy within the liver and translocation of HMGB1 from the nucleus to the cytosol in hepatocytes, effects that were inhibited by combined administration with chloroquine. In tumor cells, chloroquine increased autophagic vacuoles and LC3-II levels inhibited oxidative phosphorylation and ATP production and promoted apoptosis, which was associated with increased Annexin-V(+)/propidium iodide (PI)(-) cells, cleaved PARP, cleaved caspase-3, and cytochrome c release from mitochondria. Taken together, our findings provide a novel clinical strategy to enhance the efficacy of HDIL-2 immunotherapy for patients with cancer.
Collapse
Affiliation(s)
- Xiaoyan Liang
- Department of Surgery, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh PA, 15213
| | - Michael E. De Vera
- Department of Surgery, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh PA, 15213
| | - William J. Buchser
- Department of Surgery, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh PA, 15213
| | | | - Patricia Loughran
- Department of Surgery, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh PA, 15213
- Center for Biologic Imaging, University of Pittsburgh
| | | | - Per Basse
- Department of Immunology, University of Pittsburgh
| | - Tao Wang
- UPCI Molecular and Cellular Biology Program. University of Pittsburgh
| | | | - Herbert J. Zeh
- Department of Surgery, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh PA, 15213
| | - Michael T. Lotze
- Department of Surgery, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh PA, 15213
- Department of Immunology, University of Pittsburgh
| |
Collapse
|
18
|
Cho IC, Chung J. Current status of targeted therapy for advanced renal cell carcinoma. Korean J Urol 2012; 53:217-28. [PMID: 22536463 PMCID: PMC3332131 DOI: 10.4111/kju.2012.53.4.217] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 03/15/2012] [Indexed: 01/19/2023] Open
Abstract
The treatment of metastatic renal cell carcinoma (mRCC) has recently evolved from being predominantly cytokine-based treatment to the use of targeted agents, which include sorafenib, sunitinib, bevacizumab (plus interferon alpha [IFN-α]), temsirolimus, everolimus, pazopanib, and most recently, axitinib. Improved understanding of the molecular pathways implicated in the pathogenesis of RCC has led to the development of specific targeted therapies for treating the disease. In Korea, it has been 5 years since targeted therapy became available for mRCC. Thus, we now have broader and better therapeutic options at hand, leading to a significantly improved prognosis for patients with mRCC. However, the treatment of mRCC remains a challenge and a major health problem. Many questions remain on the efficacy of combination treatments and on the best methods for achieving complete remission. Additional studies are needed to optimize the use of these agents by identifying those patients who would most benefit and by elucidating the best means of delivering these agents, either in combination or as sequential single agents. Furthermore, numerous ongoing research activities aim at improving the benefits of the new compounds in the metastatic situation or their application in the early phase of the disease. This review introduces what is currently known regarding the fundamental biology that underlies clear cell RCC, summarizes the clinical evidence supporting the benefits of targeted agents in mRCC treatment, discusses survival endpoints used in pivotal clinical trials, and outlines future research directions.
Collapse
Affiliation(s)
- In-Chang Cho
- Department of Urology, Center for Prostate Cancer, National Cancer Center, Goyang, Korea
| | - Jinsoo Chung
- Department of Urology, Center for Prostate Cancer, National Cancer Center, Goyang, Korea
| |
Collapse
|