4101
|
Postel-Vinay S, Aspeslagh S, Lanoy E, Robert C, Soria JC, Marabelle A. Challenges of phase 1 clinical trials evaluating immune checkpoint-targeted antibodies. Ann Oncol 2015; 27:214-24. [PMID: 26578728 DOI: 10.1093/annonc/mdv550] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Immunostimulatory monoclonal antibodies (imAbs) targeting immune checkpoint molecules are revolutionizing oncology not only regarding cancer therapeutics and clinical care, but also from a drug development point of view. A handful of first-generation molecules have been approved so far based on their tremendous efficacy, after an expedited development phase that has challenged most paradigms established in the era of conventional cytotoxic therapy and to some extent molecularly targeted agents. A huge wave of second-generation imAbs is just entering into phase 1 trials now, in monotherapy or in combination. In order to maximize their chances of success in early phase trials, and eventually for patients' benefit, their clinical development has to benefit from lessons learnt from previous imAbs phase 1 trials. MATERIALS AND METHODS We reviewed the early clinical development of anti-cytotoxic T-lymphocyte antigen 4 and anti-programmed death-1 receptor/ligand. Particularities of each agent, including safety, dose--toxicity and dose--efficacy relationships, scheduling, pharmacokinetics (PK), pharmacodynamics (PD), trial design, biomarkers, response assessment and overall drug development strategies, are described and challenged. RESULTS As opposed to conventional cytotoxic agents, dose of imAbs is not linearly associated with efficacy and toxicity. Therefore, the definition of a minimal immunologically active dose could be proposed. Traditional patient eligibility criteria might also be revisited as the toxicity profile and mechanism of toxicity--immune-related adverse events--are mostly known and their physiopathology somehow less unexpected than with molecularly targeted small molecules. Most challenging are the comprehensive investigation of complex PK and PD characteristics as well as the definition of patient selection biomarkers. Finally, the early focus on efficacy (and not only dose confirmation) in expansion cohorts challenges the traditional phase 1/2/3 drug development process. CONCLUSION Several drug development paradigms have been challenged by imAbs. Here, we discuss novel approaches for an efficient and successful drug development of these agents.
Collapse
Affiliation(s)
- S Postel-Vinay
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - S Aspeslagh
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif
| | - E Lanoy
- Biostatistics and Epidemiology Unit, Gustave-Roussy, Villejuif Inserm Unit U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif
| | - C Robert
- Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif Department of Medical Oncology, Gustave Roussy, Villejuif
| | - J-C Soria
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - A Marabelle
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Inserm Unit U1015, Gustave Roussy, Villejuif, France
| |
Collapse
|
4102
|
Zhang Y, Wu S. Novel therapy for advanced gastric cancer. World J Gastrointest Oncol 2015; 7:263-270. [PMID: 26600926 PMCID: PMC4644849 DOI: 10.4251/wjgo.v7.i11.263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/14/2015] [Accepted: 09/18/2015] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is a common lethal malignancy. Gastroesophageal junction and gastric cardia tumors are the fastest rising malignancies due to increasing prevalence of obesity and acid reflex in the United States. Traditional chemotherapy remains the main treatment with trastuzumab targeting human epidermal growth factor receptor 2 positive disease. The median overall survival (OS) is less than one year for advanced GC patients; thus, there is an urgent unmet need to develop novel therapy for GC. Although multiple targeted agents were studied, only the vascular endothelial growth factor receptor inhibitor ramucirumab was approved recently by the United States Food and Drug Administration because of its 1.4 mo OS benefit (5.2 mo vs 3.8 mo, P = 0.047) as a single agent; 2.2 mo improvement of survival (9.6 mo vs 7.4 mo, P = 0.017) when combined with paclitaxel in previously treated advanced GC patients. It is the first single agent approved for previously treated GC and the second biologic agent after trastuzumab. Even with limited success, targeted therapy may be improved by developing new biomarkers. Immune therapy is changing the paradigm of cancer treatment and is presently under active investigation for GC in clinical trials. More evidence supports GC stem cells existence and early stage studies are looking for its potential therapeutic possibilities.
Collapse
|
4103
|
Spain L, Younger E, Hatipoglu E, Larkin J. Pembrolizumab in the management of metastatic melanoma. Melanoma Manag 2015; 2:315-325. [PMID: 30190860 DOI: 10.2217/mmt.15.33] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pembrolizumab is a humanized IgG4 anti-PD-1 antibody that plays a major role in the treatment of advanced melanoma. Through blockade of PD-1, it leads to an increase in effector T-cell activity in the tumor microenvironment. Clinical trial outcomes for pembrolizumab in addition to pharmacokinetics, pharmacodynamics and safety of the compound are discussed in this article. Phase I trials have demonstrated safety and efficacy of pembrolizumab in advanced, pretreated melanoma patients. When compared with chemotherapy in a Phase II trial of ipilimumab-refractory patients, those treated with pembrolizumab showed superior progression-free survival. In addition, in the pivotal Phase III trial pembrolizumab improved overall survival compared with ipilimumab in patients naive to immune checkpoint inhibition. Pembrolizumab is well tolerated and has a favorable safety profile. Common adverse events are fatigue, rash, itching and diarrhea. Less frequent immune-related adverse events include hypothyroidism, colitis, hepatitis and pneumonitis.
Collapse
Affiliation(s)
- Lavinia Spain
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - Eugenie Younger
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - Emine Hatipoglu
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - James Larkin
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| |
Collapse
|
4104
|
Jamshidi N, Jonasch E, Zapala M, Korn RL, Brooks JD, Ljungberg B, Kuo MD. The radiogenomic risk score stratifies outcomes in a renal cell cancer phase 2 clinical trial. Eur Radiol 2015; 26:2798-807. [PMID: 26560727 DOI: 10.1007/s00330-015-4082-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 09/30/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES To characterize a radiogenomic risk score (RRS), a previously defined biomarker, and to evaluate its potential for stratifying radiological progression-free survival (rPFS) in patients with metastatic renal cell carcinoma (mRCC) undergoing pre-surgical treatment with bevacizumab. METHODOLOGY In this IRB-approved study, prospective imaging analysis of the RRS was performed on phase II clinical trial data of mRCC patients (n = 41) evaluating whether patient stratification according to the RRS resulted in groups more or less likely to have a rPFS to pre-surgical bevacizumab prior to cytoreductive nephrectomy. Survival times of RRS subgroups were analyzed using Kaplan-Meier survival analysis. RESULTS The RRS is enriched in diverse molecular processes including drug response, stress response, protein kinase regulation, and signal transduction pathways (P < 0.05). The RRS successfully stratified rPFS to bevacizumab based on pre-treatment computed tomography imaging with a median progression-free survival of 6 versus >25 months (P = 0.005) and overall survival of 25 versus >37 months in the high and low RRS groups (P = 0.03), respectively. Conventional prognostic predictors including the Motzer and Heng criteria were not predictive in this cohort (P > 0.05). CONCLUSIONS The RRS stratifies rPFS to bevacizumab in patients from a phase II clinical trial with mRCC undergoing cytoreductive nephrectomy and pre-surgical bevacizumab. KEY POINTS • The RRS SOMA stratifies patient outcomes in a phase II clinical trial. • RRS stratifies subjects into prognostic groups in a discrete or continuous fashion. • RRS is biologically enriched in diverse processes including drug response programs.
Collapse
Affiliation(s)
- Neema Jamshidi
- Department of Radiological Sciences, University of California-Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Matthew Zapala
- Department of Radiological Sciences, University of California-Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
- Department of Radiology, University of California-San Diego, San Diego, CA, USA
| | | | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Borje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umea Hospital, Umea, Sweden
| | - Michael D Kuo
- Department of Radiological Sciences, University of California-Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
4105
|
|
4106
|
Engineering high-affinity PD-1 variants for optimized immunotherapy and immuno-PET imaging. Proc Natl Acad Sci U S A 2015; 112:E6506-14. [PMID: 26604307 DOI: 10.1073/pnas.1519623112] [Citation(s) in RCA: 279] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Signaling through the immune checkpoint programmed cell death protein-1 (PD-1) enables tumor progression by dampening antitumor immune responses. Therapeutic blockade of the signaling axis between PD-1 and its ligand programmed cell death ligand-1 (PD-L1) with monoclonal antibodies has shown remarkable clinical success in the treatment of cancer. However, antibodies have inherent limitations that can curtail their efficacy in this setting, including poor tissue/tumor penetrance and detrimental Fc-effector functions that deplete immune cells. To determine if PD-1:PD-L1-directed immunotherapy could be improved with smaller, nonantibody therapeutics, we used directed evolution by yeast-surface display to engineer the PD-1 ectodomain as a high-affinity (110 pM) competitive antagonist of PD-L1. In contrast to anti-PD-L1 monoclonal antibodies, high-affinity PD-1 demonstrated superior tumor penetration without inducing depletion of peripheral effector T cells. Consistent with these advantages, in syngeneic CT26 tumor models, high-affinity PD-1 was effective in treating both small (50 mm(3)) and large tumors (150 mm(3)), whereas the activity of anti-PD-L1 antibodies was completely abrogated against large tumors. Furthermore, we found that high-affinity PD-1 could be radiolabeled and applied as a PET imaging tracer to efficiently distinguish between PD-L1-positive and PD-L1-negative tumors in living mice, providing an alternative to invasive biopsy and histological analysis. These results thus highlight the favorable pharmacology of small, nonantibody therapeutics for enhanced cancer immunotherapy and immune diagnostics.
Collapse
|
4107
|
Predictive biomarkers in PD-1/PD-L1 checkpoint blockade immunotherapy. Cancer Treat Rev 2015; 41:868-76. [PMID: 26589760 DOI: 10.1016/j.ctrv.2015.11.001] [Citation(s) in RCA: 307] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 10/29/2015] [Accepted: 11/01/2015] [Indexed: 12/30/2022]
Abstract
Checkpoint blockades turn on a new paradigm shift in immunotherapy for cancer. Remarkable clinical efficacy, durable response and low toxicity of programmed death 1 (PD-1)/programmed death ligand-1 (PD-L1) checkpoint blockades have been observed in various malignancies. However, a lot of cancer patients failed to respond to the PD-1/PD-L1 checkpoint blockades. It is crucial to identify a biomarker to predict the response to checkpoint blockades. The overexpression of PD-L1 is an important and widely-explored predictive biomarker for the response to PD-1/PD-L1 antibodies. However PD-L1 staining cannot be used to accurately select patients for PD-1/PD-L1 pathway blockade due to the low prediction accuracy and dynamic changes. Tumor-infiltrating immune cells and molecules in the tumor microenvironment, or along with PD-L1 expression, may be important in predicting clinical benefits of PD-1/PD-L1 checkpoint blockades. Gene analysis has proven to be new approach for judging the potential clinical benefit of immune checkpoint inhibitors, such as mutational landscape and mismatch-repair deficiency. Further preclinical and clinical studies are necessary to carry out before its application in clinical practice. Challenges should be overcome to identify patients accurately who will benefit from PD-1/PD-L1 checkpoint blockades. In this review, we focus on the predictive biomarkers for checkpoint blockades of PD-1/PD-L1 pathway.
Collapse
|
4108
|
Zhu L, Jing S, Wang B, Wu K, Shenglin MA, Zhang S. Anti-PD-1/PD-L1 Therapy as a Promising Option for Non-Small Cell Lung Cancer: a Single arm Meta-Analysis. Pathol Oncol Res 2015; 22:331-9. [DOI: 10.1007/s12253-015-0011-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 11/04/2015] [Indexed: 01/13/2023]
|
4109
|
Zimmer L, Eigentler TK, Kiecker F, Simon J, Utikal J, Mohr P, Berking C, Kämpgen E, Dippel E, Stadler R, Hauschild A, Fluck M, Terheyden P, Rompel R, Loquai C, Assi Z, Garbe C, Schadendorf D. Open-label, multicenter, single-arm phase II DeCOG-study of ipilimumab in pretreated patients with different subtypes of metastatic melanoma. J Transl Med 2015; 13:351. [PMID: 26541511 PMCID: PMC4635983 DOI: 10.1186/s12967-015-0716-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/26/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Ipilimumab is an approved immunotherapy that has shown an overall survival benefit in patients with cutaneous metastatic melanoma in two phase III trials. As results of registrational trials might not answer all questions regarding safety and efficacy of ipilimumab in patients with advanced melanoma seen in daily clinical practice, the Dermatologic Cooperative Oncology Group conducted a phase II study to assess the efficacy and safety of ipilimumab in patients with different subtypes of metastatic melanoma. PATIENTS AND METHODS We undertook a multicenter phase II study in melanoma patients irrespective of location of the primary melanoma. Here we present data on patients with pretreated metastatic cutaneous, mucosal and occult melanoma who received up to four cycles of ipilimumab administered at a dose of 3 mg/kg in 3 week intervals. Tumor assessments were conducted at baseline, weeks 12, 24, 36 and 48 according to RECIST 1.1 criteria. Adverse events (AEs), including immune-related AEs were graded according to National Cancer Institute Common Toxicity Criteria (CTC) v.4.0. Primary endpoint was the OS rate at 12 months. RESULTS 103 pretreated patients received at least one dose of ipilimumab, including 83 cutaneous, seven mucosal and 13 occult melanomas. 1-year OS rates for cutaneous, mucosal and occult melanoma were 38 %, 14 % and 27 %, respectively. Median OS was 6.8 months (95 % CI 5.3-9.9) for cutaneous, 9.6 months (95 % CI 1.6-11.1) for mucosal, and 9.9 months (lower 95 % CI 2.3, upper 95 % CI non-existent) for occult melanoma. Overall response rates for cutaneous, mucosal and occult melanoma were 16 %, 17 % and 11 %, respectively. Eleven patients had partial response (16 %) and ten patients experienced stable disease (14 %), none achieved a complete response. Treatment-related AEs were observed in 71 patients (69 %), including 20 grade 3-4 events (19 %). No new and unexpected safety findings were noted. CONCLUSIONS Ipilimumab is a treatment option for pretreated patients with advanced cutaneous melanoma seen in daily routine. Toxicity was manageable when treated as per protocol-specific guidelines. TRIAL REGISTRATION Clinical Trials.gov NCT01355120.
Collapse
Affiliation(s)
- Lisa Zimmer
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany.
| | - Thomas K Eigentler
- Department of Dermatology, Center for Dermatooncology, University Medical Center Tübingen, Tübingen, Germany.
| | - Felix Kiecker
- Department of Dermatology and Allergy, Skin Cancer Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Jan Simon
- Department of Dermatology, Venereology and Allergology, University Hospital Leipzig, Leipzig, Germany.
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.
| | - Peter Mohr
- Department of Dermatology, Elbekliniken Stade Buxtehude, Buxtehude, Germany.
| | - Carola Berking
- Department of Dermatology and Allergy, Ludwig-Maximilian University, Munich, Germany.
| | - Eckhart Kämpgen
- Department of Dermatology, Dermatologikum Berlin, Berlin, Germany.
| | - Edgar Dippel
- Department of Dermatology, Klinikum Ludwigshafen, Skin Cancer Center Rheinpfalz, Ludwigshafen, Germany.
| | - Rudolf Stadler
- Department of Dermatology, Medical Centre Minden, Minden, Germany.
| | | | - Michael Fluck
- Department of Dermatology Hornheide, Münster, Germany.
| | | | - Rainer Rompel
- Department of Dermatology, Clinical Centre Kassel, Kassel, Germany.
| | - Carmen Loquai
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany.
| | - Zeinab Assi
- Department of Dermatology, Center for Dermatooncology, University Medical Center Tübingen, Tübingen, Germany.
| | - Claus Garbe
- Department of Dermatology, Center for Dermatooncology, University Medical Center Tübingen, Tübingen, Germany.
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
4110
|
Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, Castellano D, Choueiri TK, Gurney H, Donskov F, Bono P, Wagstaff J, Gauler TC, Ueda T, Tomita Y, Schutz FA, Kollmannsberger C, Larkin J, Ravaud A, Simon JS, Xu LA, Waxman IM, Sharma P, on behalf of the CheckMate 025 investigators.. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 2015; 373:1803-13. [PMID: 26406148 PMCID: PMC5719487 DOI: 10.1056/nejmoa1510665] [Citation(s) in RCA: 4561] [Impact Index Per Article: 456.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Nivolumab, a programmed death 1 (PD-1) checkpoint inhibitor, was associated with encouraging overall survival in uncontrolled studies involving previously treated patients with advanced renal-cell carcinoma. This randomized, open-label, phase 3 study compared nivolumab with everolimus in patients with renal-cell carcinoma who had received previous treatment. METHODS A total of 821 patients with advanced clear-cell renal-cell carcinoma for which they had received previous treatment with one or two regimens of antiangiogenic therapy were randomly assigned (in a 1:1 ratio) to receive 3 mg of nivolumab per kilogram of body weight intravenously every 2 weeks or a 10-mg everolimus tablet orally once daily. The primary end point was overall survival. The secondary end points included the objective response rate and safety. RESULTS The median overall survival was 25.0 months (95% confidence interval [CI], 21.8 to not estimable) with nivolumab and 19.6 months (95% CI, 17.6 to 23.1) with everolimus. The hazard ratio for death with nivolumab versus everolimus was 0.73 (98.5% CI, 0.57 to 0.93; P=0.002), which met the prespecified criterion for superiority (P≤0.0148). The objective response rate was greater with nivolumab than with everolimus (25% vs. 5%; odds ratio, 5.98 [95% CI, 3.68 to 9.72]; P<0.001). The median progression-free survival was 4.6 months (95% CI, 3.7 to 5.4) with nivolumab and 4.4 months (95% CI, 3.7 to 5.5) with everolimus (hazard ratio, 0.88; 95% CI, 0.75 to 1.03; P=0.11). Grade 3 or 4 treatment-related adverse events occurred in 19% of the patients receiving nivolumab and in 37% of the patients receiving everolimus; the most common event with nivolumab was fatigue (in 2% of the patients), and the most common event with everolimus was anemia (in 8%). CONCLUSIONS Among patients with previously treated advanced renal-cell carcinoma, overall survival was longer and fewer grade 3 or 4 adverse events occurred with nivolumab than with everolimus. (Funded by Bristol-Myers Squibb; CheckMate 025 ClinicalTrials.gov number, NCT01668784.).
Collapse
Affiliation(s)
| | | | | | | | - Hans J. Hammers
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | | | - Scott S. Tykodi
- University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | | | | | - Toni K. Choueiri
- Dana-Farber Cancer Institute Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA
| | - Howard Gurney
- Westmead Hospital and Macquarie University, Sydney, Australia
| | | | - Petri Bono
- Helsinki University Central Hospital, Helsinki, Finland, and University of Helsinki, Helsinki, Finland
| | - John Wagstaff
- South West Wales Cancer Institute and Swansea University College of Medicine, Swansea, UK
| | - Thomas C. Gauler
- University Hospital Essen of University of Duisburg-Essen, Germany
| | | | | | - Fabio A. Schutz
- Hospital Sao Jose, Beneficencia Portuguesa de Sao Paulo, Sao Paulo, Brazil
| | | | | | - Alain Ravaud
- Bordeaux University Hospital, Hôpital Saint André, Bordeaux, France
| | | | - Li-An Xu
- Bristol-Myers Squibb, Hopewell, NJ
| | | | | | | |
Collapse
|
4111
|
Riesenberg S, Groetchen A, Siddaway R, Bald T, Reinhardt J, Smorra D, Kohlmeyer J, Renn M, Phung B, Aymans P, Schmidt T, Hornung V, Davidson I, Goding CR, Jönsson G, Landsberg J, Tüting T, Hölzel M. MITF and c-Jun antagonism interconnects melanoma dedifferentiation with pro-inflammatory cytokine responsiveness and myeloid cell recruitment. Nat Commun 2015; 6:8755. [PMID: 26530832 PMCID: PMC4659938 DOI: 10.1038/ncomms9755] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/28/2015] [Indexed: 12/12/2022] Open
Abstract
Inflammation promotes phenotypic plasticity in melanoma, a source of non-genetic heterogeneity, but the molecular framework is poorly understood. Here we use functional genomic approaches and identify a reciprocal antagonism between the melanocyte lineage transcription factor MITF and c-Jun, which interconnects inflammation-induced dedifferentiation with pro-inflammatory cytokine responsiveness of melanoma cells favouring myeloid cell recruitment. We show that pro-inflammatory cytokines such as TNF-α instigate gradual suppression of MITF expression through c-Jun. MITF itself binds to the c-Jun regulatory genomic region and its reduction increases c-Jun expression that in turn amplifies TNF-stimulated cytokine expression with further MITF suppression. This feed-forward mechanism turns poor peak-like transcriptional responses to TNF-α into progressive and persistent cytokine and chemokine induction. Consistently, inflammatory MITF(low)/c-Jun(high) syngeneic mouse melanomas recruit myeloid immune cells into the tumour microenvironment as recapitulated by their human counterparts. Our study suggests myeloid cell-directed therapies may be useful for MITF(low)/c-Jun(high) melanomas to counteract their growth-promoting and immunosuppressive functions.
Collapse
Affiliation(s)
- Stefanie Riesenberg
- Unit for RNA Biology, Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Angela Groetchen
- Unit for RNA Biology, Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Robert Siddaway
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Tobias Bald
- Laboratory for Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Julia Reinhardt
- Unit for RNA Biology, Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Denise Smorra
- Unit for RNA Biology, Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Judith Kohlmeyer
- Laboratory for Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Marcel Renn
- Laboratory for Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Bengt Phung
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Barngatan 2B, Lund 221 85, Sweden
| | - Pia Aymans
- Laboratory for Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Tobias Schmidt
- Institute of Molecular Medicine, University Hospital, University of Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Veit Hornung
- Institute of Molecular Medicine, University Hospital, University of Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Irwin Davidson
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, 1 Rue Laurent Fries, Illkirch Cédex 67404, France
| | - Colin R Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Göran Jönsson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Barngatan 2B, Lund 221 85, Sweden
| | - Jennifer Landsberg
- Laboratory for Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Thomas Tüting
- Laboratory for Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | - Michael Hölzel
- Unit for RNA Biology, Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| |
Collapse
|
4112
|
Fakhrejahani F, Tomita Y, Maj-Hes A, Trepel JB, De Santis M, Apolo AB. Immunotherapies for bladder cancer: a new hope. Curr Opin Urol 2015; 25:586-96. [PMID: 26372038 PMCID: PMC6777558 DOI: 10.1097/mou.0000000000000213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We review recent data on immunotherapies for bladder cancer and discuss strategies to maximize the antitumor effect of immunotherapy in solid tumors. RECENT FINDINGS Anti-programmed death ligand 1 has shown promise in advanced bladder cancer. Clinical trials of immune checkpoint inhibitors as monotherapy or in combination are underway. Here we review strategies for enhancing antitumor immunity using immunomodulating agents or combination treatments that may increase tumor response. SUMMARY Combining immune checkpoint inhibitors with other treatment modalities may lead to the development of new treatment strategies in advanced bladder cancer; however, identifying predictive biomarkers is essential for appropriate patient selection.
Collapse
Affiliation(s)
- Farhad Fakhrejahani
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yusuke Tomita
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Agnes Maj-Hes
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria De Santis
- Cancer Research Unit, Warwick University Medical School, Coventry, UK
| | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
4113
|
Hanna GG, Coyle VM, Prise KM. Immune modulation in advanced radiotherapies: Targeting out-of-field effects. Cancer Lett 2015; 368:246-51. [DOI: 10.1016/j.canlet.2015.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/03/2015] [Accepted: 04/08/2015] [Indexed: 01/09/2023]
|
4114
|
Cherington CC, Bryce AH. The end of the beginning: PD-1 inhibition as the new standard of care first-line immunotherapy in metastatic melanoma. Melanoma Manag 2015; 2:305-309. [PMID: 30190858 PMCID: PMC6094642 DOI: 10.2217/mmt.15.32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Chad C Cherington
- Department of Medical Oncology, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA
| | - Alan H Bryce
- Department of Medical Oncology, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA
| |
Collapse
|
4115
|
Munhoz RR, González AF, Reed VA, Postow MA. Targeting immune checkpoints in melanoma: an update. Melanoma Manag 2015; 2:339-352. [PMID: 30190862 PMCID: PMC6094685 DOI: 10.2217/mmt.15.25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Different treatment modalities encompassed under the term 'immunotherapy' have led to major breakthroughs in the treatment of melanoma. Immune checkpoint-blocking antibodies targeting CTLA-4 and PD-1 result in significant activity and prolonged survival in patients with advanced melanoma and are currently available for clinical use. Studies addressing novel immune checkpoint blocking antibodies, combined approaches and predictive/prognostic biomarkers are expected to broaden the applicability and efficacy of this approach. In this article, we will review clinically meaningful aspects of immune checkpoint blockade, promising strategies under development and the challenges faced in a continuous search to improve the outcomes of patients affected by this disease.
Collapse
Affiliation(s)
- Rodrigo R Munhoz
- Melanoma & Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo, Brazil
| | | | - Vanessa A Reed
- Melanoma & Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael A Postow
- Melanoma & Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
4116
|
Hersh EM, Del Vecchio M, Brown MP, Kefford R, Loquai C, Testori A, Bhatia S, Gutzmer R, Conry R, Haydon A, Robert C, Ernst S, Homsi J, Grob JJ, Kendra K, Agarwala SS, Li M, Clawson A, Brachmann C, Karnoub M, Elias I, Renschler MF, Hauschild A. A randomized, controlled phase III trial of nab-Paclitaxel versus dacarbazine in chemotherapy-naïve patients with metastatic melanoma. Ann Oncol 2015; 26:2267-74. [PMID: 26410620 PMCID: PMC6279094 DOI: 10.1093/annonc/mdv324] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 05/08/2015] [Accepted: 07/21/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The efficacy and safety of nab-paclitaxel versus dacarbazine in patients with metastatic melanoma was evaluated in a phase III randomized, controlled trial. PATIENTS AND METHODS Chemotherapy-naïve patients with stage IV melanoma received nab-paclitaxel 150 mg/m(2) on days 1, 8, and 15 every 4 weeks or dacarbazine 1000 mg/m(2) every 3 weeks. The primary end point was progression-free survival (PFS) by independent radiologic review; the secondary end point was overall survival (OS). RESULTS A total of 529 patients were randomized to nab-paclitaxel (n = 264) or dacarbazine (n = 265). Baseline characteristics were well balanced. The majority of patients were men (66%), had an Eastern Cooperative Oncology Group status of 0 (71%), and had M1c stage disease (65%). The median PFS (primary end point) was 4.8 months with nab-paclitaxel and 2.5 months with dacarbazine [hazard ratio (HR), 0.792; 95.1% confidence interval (CI) 0.631-0.992; P = 0.044]. The median OS was 12.6 months with nab-paclitaxel and 10.5 months with dacarbazine (HR, 0.897; 95.1% CI 0.738-1.089; P = 0.271). Independently assessed overall response rate was 15% versus 11% (P = 0.239), and disease control rate (DCR) was 39% versus 27% (P = 0.004) for nab-paclitaxel versus dacarbazine, respectively. The most common grade ≥3 treatment-related adverse events were neuropathy (nab-paclitaxel, 25% versus dacarbazine, 0%; P < 0.001), and neutropenia (nab-paclitaxel, 20% versus dacarbazine, 10%; P = 0.004). There was no correlation between secreted protein acidic and rich in cysteine (SPARC) status and PFS in either treatment arm. CONCLUSIONS nab-Paclitaxel significantly improved PFS and DCR compared with dacarbazine, with a manageable safety profile.
Collapse
Affiliation(s)
- E M Hersh
- Department of Medicine, Arizona Cancer Center, Tucson, USA
| | - M Del Vecchio
- Department of Medical Oncology, Fondazione IRCCS National Tumor Institute, Milan, Italy
| | - M P Brown
- Cancer Clinical Trials Unit, Royal Adelaide Hospital and School of Medicine, University of Adelaide, Adelaide
| | - R Kefford
- Sydney West Cancer Trials Centre/Westmead Hospital and Melanoma Institute Australia, University of Sydney, North Sydney, Australia
| | - C Loquai
- Department of Dermatology, University of Mainz, Mainz, Germany
| | - A Testori
- Melanoma and Muscle Cutaneous Sarcoma Division, European Institute of Oncology, Milan, Italy
| | - S Bhatia
- Department of Medicine, Seattle Cancer Care Alliance, Seattle, USA
| | - R Gutzmer
- Department of Dermatology and Oncology, Hannover Medical School, Hannover, Germany
| | - R Conry
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, USA
| | - A Haydon
- Department of Medical Oncology, Alfred Hospital, Melbourne, Australia
| | - C Robert
- Demartology Unit, Department of Medicine, The Gustave Roussy Cancer Institute, Villejuif, France
| | - S Ernst
- Department of Medical Oncology, London Health Sciences Center-London Regional Cancer Program, London, Canada
| | - J Homsi
- Department of Medical Oncology, Banner MD Anderson Cancer Center, Gilbert, USA
| | - J J Grob
- Department of Dermatology, Timone Hospital, APHM and Aix-Marseille University, Marseille, France
| | - K Kendra
- Department of Internal Medicine, Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus
| | - S S Agarwala
- Department of Hematology and Oncology, St Luke's Cancer Center and Temple University, Bethlehem
| | - M Li
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - A Clawson
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - C Brachmann
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - M Karnoub
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - I Elias
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - M F Renschler
- Biometrics and Data Operations/Translational Medicine/Biometrics and Data Operations/Clinical Research & Development/Global Medical Affairs, Celgene Corporation, Summit, USA
| | - A Hauschild
- Department of Dermatology, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
4117
|
Tanese K, Hashimoto Y, Berkova Z, Wang Y, Samaniego F, Lee JE, Ekmekcioglu S, Grimm EA. Cell Surface CD74-MIF Interactions Drive Melanoma Survival in Response to Interferon-γ. J Invest Dermatol 2015; 135:2775-2784. [PMID: 26039541 PMCID: PMC4640965 DOI: 10.1038/jid.2015.204] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 04/06/2015] [Accepted: 05/03/2015] [Indexed: 12/31/2022]
Abstract
Melanoma is believed to be a highly immunogenic tumor and recent developments in immunotherapies are promising. IFN-γ produced by immune cells has a crucial role in tumor immune surveillance; however, it has also been reported to be pro-tumorigenic. In the current study, we found that IFN-γ enhances the expression of CD74, which interacts with its ligand, macrophage migration inhibitory factor (MIF), and thereby activates the PI3K/AKT pathway in melanoma, promoting tumor survival. IFN-γ increased phosphorylation of AKT Ser473 and upregulated total cell surface expression of CD74 in human melanoma cell lines tested. CD74 was highly expressed in melanoma tissues. Moreover, the expression of CD74 on tumor cells correlated with plasma IFN-γ levels in melanoma patient samples. In our analysis of melanoma cell lines, all produced MIF constitutively. Blockade of CD74-MIF interaction reduced AKT phosphorylation and expression of pro-tumorigenic molecules, including IL-6, IL-8, and BCL-2. Inhibition of CD74-MIF interaction significantly suppressed tumor growth in the presence of IFN-γ in our xenograft mouse model. Thus, we conclude that IFN-γ promotes melanoma cell survival by regulating CD74-MIF signaling, suggesting that targeting the CD74-MIF interaction under IFN-γ-stimulatory conditions would be an effective therapeutic approach for melanoma.
Collapse
Affiliation(s)
- Keiji Tanese
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Yuuri Hashimoto
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zuzana Berkova
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuling Wang
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Felipe Samaniego
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Elizabeth A Grimm
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
4118
|
Lee SJ, Kim TM, Kim YJ, Jang KT, Lee HJ, Lee SN, Ahn MS, Hwang IG, Lee S, Lee MH, Lee J. Phase II Trial of Nilotinib in Patients With Metastatic Malignant Melanoma Harboring KIT Gene Aberration: A Multicenter Trial of Korean Cancer Study Group (UN10-06). Oncologist 2015; 20:1312-1319. [PMID: 26424760 PMCID: PMC4718426 DOI: 10.1634/theoncologist.2015-0161] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/18/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND KIT has been suggested to be a potential therapeutic target for malignant melanoma. We evaluated the antitumor activity and safety of the KIT inhibitor nilotinib in metastatic melanoma patients harboring KIT gene mutations or amplifications. METHODS We conducted a phase II multicenter trial of nilotinib in metastatic malignant melanoma with KIT mutations or amplifications. Patients received 400 mg oral nilotinib twice daily. The primary endpoint was response rate, and if seven or more responders were observed from the cumulative 36 patients, nilotinib would be considered worthy of further testing in this study population. RESULTS Between October 2009 and June 2013, 176 patients underwent molecular screening for KIT gene aberrations, and 42 patients harboring KIT gene mutations and/or amplification were enrolled in the study. Overall, 25 (59.5%), 15 (35.7%), and 2 (4.8%) patients had KIT mutations, KIT amplifications, and both KIT mutations and amplification, respectively. Of the 42 enrolled patients, 1 patient achieved complete response, 6 patients achieved partial response, and 17 patients achieved stable disease, resulting in an overall response rate of 16.7% (95% confidence interval [CI]: 5.4%-28.0%) and a disease control rate of 57.1% (95% CI: 42.1%-72.1%). The median duration of response was 34 weeks (range: 5-55 weeks). Of the 7 responders, 6 patients had KIT mutations (exon 11: 5 patients; exon 17: 1 patient), and 1 patient had KIT amplification only. CONCLUSION Although this study did not meet its primary endpoint of response rate, nilotinib showed durable response in a subset of metastatic melanoma patients with specific KIT mutations. IMPLICATIONS FOR PRACTICE KIT aberration can be detected in a subset of metastatic melanoma patients. This phase II trial showed that nilotinib demonstrates durable response in a subset of patients with KIT mutations. The safety profile was very tolerable. This study suggests that a KIT inhibitor may benefit a small subset of metastatic melanoma patients with KIT mutations.
Collapse
Affiliation(s)
- Su Jin Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Jung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Kee-Taek Jang
- Department of Pathology and Translational Genomics, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyo Jin Lee
- Division of Hematology/Oncology, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Soon Nam Lee
- Division of Hemato-Oncology, Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Mi Sun Ahn
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - In Gyu Hwang
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Suee Lee
- Department of Internal Medicine, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Moon-Hee Lee
- Department of Hemato-Oncology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Jeeyun Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
4119
|
Okada H, Weller M, Huang R, Finocchiaro G, Gilbert MR, Wick W, Ellingson BM, Hashimoto N, Pollack IF, Brandes AA, Franceschi E, Herold-Mende C, Nayak L, Panigrahy A, Pope WB, Prins R, Sampson JH, Wen PY, Reardon DA. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol 2015; 16:e534-e542. [PMID: 26545842 PMCID: PMC4638131 DOI: 10.1016/s1470-2045(15)00088-1] [Citation(s) in RCA: 542] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/13/2015] [Accepted: 06/15/2015] [Indexed: 12/14/2022]
Abstract
Immunotherapy is a promising area of therapy in patients with neuro-oncological malignancies. However, early-phase studies show unique challenges associated with the assessment of radiological changes in response to immunotherapy reflecting delayed responses or therapy-induced inflammation. Clinical benefit, including long-term survival and tumour regression, can still occur after initial disease progression or after the appearance of new lesions. Refinement of the response assessment criteria for patients with neuro-oncological malignancies undergoing immunotherapy is therefore warranted. Herein, a multinational and multidisciplinary panel of neuro-oncology immunotherapy experts describe immunotherapy Response Assessment for Neuro-Oncology (iRANO) criteria based on guidance for the determination of tumour progression outlined by the immune-related response criteria and the RANO working group. Among patients who demonstrate imaging findings meeting RANO criteria for progressive disease within 6 months of initiating immunotherapy, including the development of new lesions, confirmation of radiographic progression on follow-up imaging is recommended provided that the patient is not significantly worse clinically. The proposed criteria also include guidelines for the use of corticosteroids. We review the role of advanced imaging techniques and the role of measurement of clinical benefit endpoints including neurological and immunological functions. The iRANO guidelines put forth in this Review will evolve successively to improve their usefulness as further experience from immunotherapy trials in neuro-oncology accumulate.
Collapse
Affiliation(s)
- Hideho Okada
- Department of Neurological Surgery, University of California, San
Francisco, San Francisco, CA, USA
| | - Michael Weller
- Department of Neurology, University Hospital Zurich, Zurich,
Switzerland
| | - Raymond Huang
- Department of Radiology, Brigham and Women's Hospital, Boston,
MA, USA
| | | | - Mark R. Gilbert
- Neuro-Oncology Branch, National Institutes of Health, Bethesda,
MD, USA
| | - Wolfgang Wick
- Department of Neurooncology, Heidelberg University Hospital,
Heidelberg, Germany
| | - Benjamin M. Ellingson
- Departments of Radiological Sciences, Bioengineering, Biomedical
Physics, and Psychiatry David Geffen School of Medicine University of California, Los
Angeles Los Angeles, CA, USA
| | - Naoya Hashimoto
- Department of Neurosurgery, Osaka University Graduate School of
Medicine, Suita, Osaka, Japan
| | - Ian F. Pollack
- Department of Neurological Surgery, University of Pittsburgh
School of Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Alba A. Brandes
- Department of Medical Oncology, Azienda USL–IRCCS
Institute of Neurological Science, Bologna, Italy
| | - Enrico Franceschi
- Department of Medical Oncology, Azienda USL–IRCCS
Institute of Neurological Science, Bologna, Italy
| | - Christel Herold-Mende
- Department of Neurosurgery, Division of Experimental
Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Lakshmi Nayak
- Center for Neuro-Oncology, Dana-Farber Cancer Institute,
Boston, MA, USA
| | - Ashok Panigrahy
- Department of Radiology, University of Pittsburgh School of
Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Whitney B. Pope
- Department of Radiology, David Geffen School of Medicine at
University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert Prins
- Department of Neurosurgery, David Geffen School of Medicine at
University of California, Los Angeles, Los Angeles, CA, USA
| | - John H. Sampson
- Department of Neurosurgery, Duke University School of Medicine,
Durham, NC, USA
| | - Patrick Y. Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute,
Boston, MA, USA
| | - David A. Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute,
Boston, MA, USA
| |
Collapse
|
4120
|
Eroglu Z, Kim DW, Wang X, Camacho LH, Chmielowski B, Seja E, Villanueva A, Ruchalski K, Glaspy JA, Kim KB, Hwu WJ, Ribas A. Long term survival with cytotoxic T lymphocyte-associated antigen 4 blockade using tremelimumab. Eur J Cancer 2015; 51:2689-97. [PMID: 26364516 PMCID: PMC4821004 DOI: 10.1016/j.ejca.2015.08.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 06/25/2015] [Accepted: 08/14/2015] [Indexed: 11/23/2022]
Abstract
PURPOSE One of the hallmarks of cancer immunotherapy is the long duration of responses, evident with cytokines like interleukin-2 or a variety of cancer vaccines. However, there is limited information available on very long term outcomes of patients treated with anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies. Tremelimumab is an anti-CTLA-4 antibody of immunoglobulin G2 (IgG2) isotype initially tested in patients with advanced melanoma over 12 years ago. METHODS We reviewed the outcomes of patients with advanced melanoma enrolled in four phase 1 and 2 tremelimumab trials at two sites to determine response rates and long-term survival. RESULTS A total of 143 patients were enrolled at two institutions from 2002 to 2008. Tremelimumab administration varied between a single dose of 0.01 mg/kg and 15 mg/kg every 3 months. Median overall survival was 13 months (95% confidence interval (CI), 10-16.6), ranging from less than a month to 12+ years. An objective response rate of 15.6% was observed, with median duration of response of 6.5 years, range of 3-136+ months. The Kaplan-Meier estimated 5 year survival rate was 20% (95% CI, 13-26%), with 10 and 12.5 year survival rates of 16% (95% CI, 9-23%). CONCLUSIONS CTLA-4 blockade with tremelimumab can lead to very long duration of objective anti-tumour responses beyond 12 years.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/immunology
- Antineoplastic Agents/therapeutic use
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Humans
- Immunotherapy/methods
- Kaplan-Meier Estimate
- Male
- Melanoma/immunology
- Melanoma/mortality
- Melanoma/therapy
- Middle Aged
- Outcome Assessment, Health Care/methods
- Outcome Assessment, Health Care/statistics & numerical data
- Remission Induction
- Retrospective Studies
- Survival Rate
- Survivors/statistics & numerical data
- Time Factors
- Young Adult
Collapse
Affiliation(s)
- Zeynep Eroglu
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Dae Won Kim
- Department of Melanoma Medical Oncology, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoyan Wang
- Department of Medicine Statistics Core, University of California Los Angeles, Los Angeles, CA, USA
| | - Luis H Camacho
- St. Luke's Medical Center Cancer Center, Houston, TX, USA
| | - Bartosz Chmielowski
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Elizabeth Seja
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Arturo Villanueva
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Kathleen Ruchalski
- Department of Radiology, University of California Los Angeles, Los Angeles, CA, USA
| | - John A Glaspy
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Kevin B Kim
- Department of Melanoma Medical Oncology, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA; California Pacific Medical Center, San Francisco, CA, USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
4121
|
Prognostic Nutritional Index Predicts Severe Complications, Recurrence, and Poor Prognosis in Patients With Colorectal Cancer Undergoing Primary Tumor Resection. Dis Colon Rectum 2015; 58:1048-57. [PMID: 26445177 DOI: 10.1097/dcr.0000000000000458] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The prognostic nutritional index is reportedly related to postoperative outcomes. OBJECTIVE The aim of this study was to elucidate the clinical importance of the prognostic nutritional index in patients with colorectal cancer who were undergoing primary tumor resection. DESIGN This is a retrospective study from a single institution. SETTINGS This study was conducted at a colorectal surgery service in an academic teaching hospital. PATIENTS The 556 patients with colorectal cancer who were undergoing surgery between March 2005 and August 2014 were eligible for this study. MAIN OUTCOME MEASURES The preoperative prognostic nutritional index was calculated. Classification and regression tree analysis was performed to determine the prognostic nutritional index cutoff value. The associations of the prognostic nutritional index status with clinicopathological factors and postoperative outcomes were examined using univariate and multivariate analyses. RESULTS Classification and regression tree analysis demonstrated that 45.5 was the optimal cutoff value. The low status (≤45.5) was correlated with older age, low BMI, low estimated glomerular filtration rate, CEA positivity, carbohydrate antigen 19-9 positivity, preoperative chemotherapy, tumors invading muscular or deeper layers, distant metastasis, poor differentiation, severe postoperative complications, tumor recurrence, and poor survival. In multivariate analysis, the low status was an independent risk factor for severe postoperative complications (OR = 2.06 [95% CI, 1.22-3.50]; p = 0.007) and low overall survival (HR =3.98 [95% CI, 2.38-6.89]; p < 0.001). LIMITATIONS Our data set was collected retrospectively from a single institution. In addition, our study was only for preoperative prognostic nutritional index status, not considering the postoperative host status. CONCLUSIONS The preoperative prognostic nutritional index predicts severe complications, recurrence, and poor prognosis in patients with colorectal cancer who are undergoing primary tumor resection. Investigation of the nutritional and immunologic statuses using the prognostic nutritional index could be a useful clinical approach.
Collapse
|
4122
|
Tsai KK, Daud AI. Nivolumab plus ipilimumab in the treatment of advanced melanoma. J Hematol Oncol 2015; 8:123. [PMID: 26518223 PMCID: PMC4628394 DOI: 10.1186/s13045-015-0219-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/13/2015] [Indexed: 12/17/2022] Open
Abstract
Advanced melanoma has historically been a difficult disease to treat due to few effective systemic treatment options. However, over the past few years, scientific advancements in immune checkpoint inhibition have resulted in several novel approaches that have changed front-line management of advanced melanoma. Despite these exciting developments, there remains room for improvement in treatment outcomes. Combination immunotherapy, in particular combined cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed death 1 (PD-1) blockade, represents an important first step in this direction.
Collapse
Affiliation(s)
- Katy K Tsai
- Helen Diller Comprehensive Cancer Center, University of California, 1600 Divisadero St., Box 1770, San Francisco, CA, 94115, USA.
| | - Adil I Daud
- Helen Diller Comprehensive Cancer Center, University of California, 1600 Divisadero St., Box 1770, San Francisco, CA, 94115, USA.
| |
Collapse
|
4123
|
Regulatory T cells in the immunotherapy of melanoma. Tumour Biol 2015; 37:77-85. [PMID: 26515336 DOI: 10.1007/s13277-015-4315-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022] Open
Abstract
Patients with melanoma are supposed to develop spontaneous immune responses against specific tumor antigens. However, several mechanisms contribute to the failure of tumor antigen-specific T cell responses, inducing immune escape. Importantly, immunosuppression mediated by regulatory T cells (Tregs) in tumor lesions is a dominant mechanism of tumor immune evasion. Based on this information, several therapies targeting Tregs such as cyclophosphamide, IL-2-based therapies, and antibodies against the surface molecular of Tregs have been developed. However, only some of these strategies showed clinical efficacy in patients with melanoma in spite of their success in shifting immune systems to antitumor responses in animal models. In the future, strategies specifically depleting local Tregs, inhibiting Treg migration to the tumor lesion, and Treg depletion in combination with other chemotherapies or immune modulation will hopefully bring benefits to melanoma patients.
Collapse
|
4124
|
Geukes Foppen MH, Donia M, Svane IM, Haanen JBAG. Tumor-infiltrating lymphocytes for the treatment of metastatic cancer. Mol Oncol 2015; 9:1918-35. [PMID: 26578452 DOI: 10.1016/j.molonc.2015.10.018] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/15/2022] Open
Abstract
Over the past few years melanoma incidence has been rising steadily, resulting in an increase in melanoma related mortality. Until recently, therapeutic options for metastatic melanoma were scarce. Chemotherapy and, in some countries, IL-2 were the only registered treatment modalities. In the last five years, treatment with immunotherapy (anti CTLA-4, anti PD-1, or the combination of these antibodies) has shown very promising results and was able to improve survival in patients with metastatic melanoma. Adoptive cell therapy using tumor-infiltrating lymphocytes is yet another, but highly promising, immunotherapeutic strategy for patients with metastatic melanoma. This review will discuss the development of TIL as a treatment option for melanoma, its mode of action and simplification over time, and the possibilities to expand this therapy to other types of cancer. Also, the future directions of TIL based therapies will be highlighted.
Collapse
Affiliation(s)
- M H Geukes Foppen
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| | - M Donia
- Center for Cancer Immune Therapy, Department of Haematology and Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - I M Svane
- Center for Cancer Immune Therapy, Department of Haematology and Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - J B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
4125
|
Abdel-Rahman O, ElHalawani H, Fouad M. Risk of gastrointestinal complications in cancer patients treated with immune checkpoint inhibitors: a meta-analysis. Immunotherapy 2015; 7:1213-27. [PMID: 26513491 DOI: 10.2217/imt.15.87] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM We performed a meta-analysis of the risk of selected gastrointestinal toxicities associated with immune checkpoint inhibitors. PATIENTS & METHODS Eligible studies included randomized trials of patients with solid tumors on ipilimumab, nivolumab, pembrolizumab, tremelimumab, pidilizumab and atezolizumab, describing events of diarrhea, vomiting or colitis. RESULTS After exclusion of ineligible studies, a total of ten clinical trials were considered eligible for the meta-analysis. The relative risk of all-grade diarrhea, vomiting and colitis was 1.64 (95% CI: 1.19-2.26; p = 0.002), 0.72 (95% CI: 0.49-1.07; p = 0.1), 10.35 (95% CI: 5.78-18.53; p < 0.00001), respectively. CONCLUSION Our meta-analysis has demonstrated that immune checkpoint inhibitors are associated with a significantly increased risk of all grade and high-grade colitis.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hesham ElHalawani
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mona Fouad
- Medical Microbiology & Immunology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
4126
|
Galluzzi L, Eggermont A, Kroemer G. Doubling the blockade for melanoma immunotherapy. Oncoimmunology 2015; 5:e1106127. [PMID: 26942094 DOI: 10.1080/2162402x.2015.1106127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 12/19/2022] Open
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | | | - Guido Kroemer
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
4127
|
Desrichard A, Snyder A, Chan TA. Cancer Neoantigens and Applications for Immunotherapy. Clin Cancer Res 2015; 22:807-12. [PMID: 26515495 DOI: 10.1158/1078-0432.ccr-14-3175] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/18/2015] [Indexed: 12/12/2022]
Abstract
Recent advances in immune checkpoint blockade therapy have revolutionized the treatment of cancer. Tumor-specific antigens that are generated by somatic mutation, neoantigens, can influence patient response to immunotherapy and contribute to tumor shrinkage. Recent evidence demonstrating the success of checkpoint blockade immunotherapy in boosting T-cell reactivity against patient-specific neoantigens constitutes a strong rationale for the development of personalized vaccines against these nonself peptides. With the decreasing cost of next-generation sequencing, peptide manufacturing, and improvement of in silico prediction of peptide immunogenicity, it is increasingly important to evaluate the potential use of neoantigens in both diagnosis and treatment. Specifically, these neoantigens could be useful both as predictors of immune checkpoint blockade therapy response and/or incorporated in therapeutic vaccination strategies.
Collapse
Affiliation(s)
- Alexis Desrichard
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexandra Snyder
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
4128
|
Lord JD. Promises and paradoxes of regulatory T cells in inflammatory bowel disease. World J Gastroenterol 2015; 21:11236-45. [PMID: 26523099 PMCID: PMC4616201 DOI: 10.3748/wjg.v21.i40.11236] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/02/2015] [Accepted: 08/28/2015] [Indexed: 02/06/2023] Open
Abstract
Since their discovery two decades ago, CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) have become the subject of intense investigation by immunologists. Unlike other T cells, which promote an immune response, Tregs actively inhibit inflammation when activated by their cognate antigen, thus raising hope that these cells could be engineered into a highly targeted, antigen-specific, immunosuppressant therapy. Although Tregs represent less than 10% of circulating CD4(+)T cells, they have been shown to play an essential role in preventing or limiting inflammation in a variety of animal models and human diseases. In particular, spontaneous intestinal inflammation has been shown to occur in the absence of Tregs, suggesting that there may be a Treg defect central to the pathogenesis of human inflammatory bowel disease (IBD). However, over the past decade, multiple groups have reported no qualitative or quantitative deficits in Tregs from the intestines and blood of IBD patients to explain why these cells fail to regulate inflammation in Crohn's disease and ulcerative colitis. In this review, we will discuss the history of Tregs, what is known about them in IBD, and what progress and obstacles have been seen with efforts to employ them for therapeutic benefit.
Collapse
|
4129
|
Śledzińska A, Menger L, Bergerhoff K, Peggs KS, Quezada SA. Negative immune checkpoints on T lymphocytes and their relevance to cancer immunotherapy. Mol Oncol 2015; 9:1936-65. [PMID: 26578451 DOI: 10.1016/j.molonc.2015.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 10/09/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023] Open
Abstract
The term 'inhibitory checkpoint' refers to the broad spectrum of co-receptors expressed by T cells that negatively regulate T cell activation thus playing a crucial role in maintaining peripheral self-tolerance. Co-inhibitory receptor ligands are highly expressed by a variety of malignancies allowing evasion of anti-tumour immunity. Recent studies demonstrate that manipulation of these co-inhibitory pathways can remove the immunological brakes that impede endogenous immune responses against tumours. Antibodies that block the interactions between co-inhibitory receptors and their ligands have delivered very promising clinical responses, as has been shown by recent successful trials targeting the CTLA-4 and PD-1 pathways. In this review, we discuss the mechanisms of action and expression pattern of co-inhibitory receptors on different T cells subsets, emphasising differences between CD4(+) and CD8(+) T cells. We also summarise recent clinical findings utilising immune checkpoint blockade.
Collapse
Affiliation(s)
- Anna Śledzińska
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK
| | - Laurie Menger
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK
| | | | - Karl S Peggs
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK.
| | | |
Collapse
|
4130
|
Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov 2015; 14:561-84. [PMID: 26228759 DOI: 10.1038/nrd4591] [Citation(s) in RCA: 1002] [Impact Index Per Article: 100.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Targeting immune checkpoints such as programmed cell death protein 1 (PD1), programmed cell death 1 ligand 1 (PDL1) and cytotoxic T lymphocyte antigen 4 (CTLA4) has achieved noteworthy benefit in multiple cancers by blocking immunoinhibitory signals and enabling patients to produce an effective antitumour response. Inhibitors of CTLA4, PD1 or PDL1 administered as single agents have resulted in durable tumour regression in some patients, and combinations of PD1 and CTLA4 inhibitors may enhance antitumour benefit. Numerous additional immunomodulatory pathways as well as inhibitory factors expressed or secreted by myeloid and stromal cells in the tumour microenvironment are potential targets for synergizing with immune checkpoint blockade. Given the breadth of potential targets in the immune system, critical questions to address include which combinations should move forward in development and which patients will benefit from these treatments. This Review discusses the leading drug targets that are expressed on tumour cells and in the tumour microenvironment that allow enhancement of the antitumour immune response.
Collapse
Affiliation(s)
- Kathleen M Mahoney
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA. [2] Division of Haematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA. [3]
| | - Paul D Rennert
- 1] SugarCone Biotech, Holliston, Massachusetts 01746, USA. [2] Videre Biotherapeutics, Watertown, Massachusetts 02472, USA. [3]
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
4131
|
Ettahri H, Elomrani F, Elkabous M, Rimani M, Boutayeb S, Mrabti H, Errihani H. Duodenal and gallbladder metastasis of regressive melanoma: a case report and review of the literature. J Gastrointest Oncol 2015; 6:E77-81. [PMID: 26487955 DOI: 10.3978/j.issn.2078-6891.2015.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Malignant melanoma involving the gastrointestinal (GI) tract may be primary or metastatic. Small bowel is the commonest site of GI metastases from cutaneous malignant melanoma, metastatic lesion in the gallbladder is extremely rare. CASE PRESENTATION This case report describes the presentation of metastatic melanoma in duodenum and gallbladder. A 45-year-old man has presented melena with intermittent abdominal pain. On physical examination we found a small lesion between the fourth and fifth toes, associated with inguinal lymph node. An Abdominal ultrasound revealed diffuse duodenal thickening. Upper endoscopy was performed and discovered an ulcerative lesion in the second part of the duodenum. The biopsy with immunohistochemical stains was in favor of a duodenal location of melanoma. Computed tomography (CT) revealed many circumferential thickening of ileal loops associated with a nodular lesion in the anterior wall of the gallbladder. The patient was treated by palliative chemotherapy. DISCUSSION Malignant melanoma of the GI tract may be primary or secondary. The small bowel is the most affected, but it's rare in the gallbladder. The clinical presentation can mimic the other intestinal tumors, and the diagnosis is based on imaging; CT scan and GI endoscopy have a key role on the diagnosis, and the treatment depends on the location and the number of lesions. CONCLUSIONS Metastases of melanoma in the GI tract are uncommon, the diagnosis must be suspected in any patient with a history of melanoma with digestive signs.
Collapse
Affiliation(s)
- Hamza Ettahri
- 1 Department of Medical Oncology, National Institute of Oncology, Rabat 10000, Morocco ; 2 Faculté de Médecine et de Pharmacie de Rabat, Université Mohamed V Souissi, Rabat 8007, Morocco ; 3 Centre de Pathologie Hassan, Rabat, Morocco
| | - Fadwa Elomrani
- 1 Department of Medical Oncology, National Institute of Oncology, Rabat 10000, Morocco ; 2 Faculté de Médecine et de Pharmacie de Rabat, Université Mohamed V Souissi, Rabat 8007, Morocco ; 3 Centre de Pathologie Hassan, Rabat, Morocco
| | - Mustapha Elkabous
- 1 Department of Medical Oncology, National Institute of Oncology, Rabat 10000, Morocco ; 2 Faculté de Médecine et de Pharmacie de Rabat, Université Mohamed V Souissi, Rabat 8007, Morocco ; 3 Centre de Pathologie Hassan, Rabat, Morocco
| | - Mouna Rimani
- 1 Department of Medical Oncology, National Institute of Oncology, Rabat 10000, Morocco ; 2 Faculté de Médecine et de Pharmacie de Rabat, Université Mohamed V Souissi, Rabat 8007, Morocco ; 3 Centre de Pathologie Hassan, Rabat, Morocco
| | - Saber Boutayeb
- 1 Department of Medical Oncology, National Institute of Oncology, Rabat 10000, Morocco ; 2 Faculté de Médecine et de Pharmacie de Rabat, Université Mohamed V Souissi, Rabat 8007, Morocco ; 3 Centre de Pathologie Hassan, Rabat, Morocco
| | - Hind Mrabti
- 1 Department of Medical Oncology, National Institute of Oncology, Rabat 10000, Morocco ; 2 Faculté de Médecine et de Pharmacie de Rabat, Université Mohamed V Souissi, Rabat 8007, Morocco ; 3 Centre de Pathologie Hassan, Rabat, Morocco
| | - Hassan Errihani
- 1 Department of Medical Oncology, National Institute of Oncology, Rabat 10000, Morocco ; 2 Faculté de Médecine et de Pharmacie de Rabat, Université Mohamed V Souissi, Rabat 8007, Morocco ; 3 Centre de Pathologie Hassan, Rabat, Morocco
| |
Collapse
|
4132
|
Kleponis J, Skelton R, Zheng L. Fueling the engine and releasing the break: combinational therapy of cancer vaccines and immune checkpoint inhibitors. Cancer Biol Med 2015; 12:201-8. [PMID: 26487965 PMCID: PMC4607816 DOI: 10.7497/j.issn.2095-3941.2015.0046] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors are increasingly drawing much attention in the therapeutic development for cancer treatment. However, many cancer patients do not respond to treatments with immune checkpoint inhibitors, partly because of the lack of tumor-infiltrating effector T cells. Cancer vaccines may prime patients for treatments with immune checkpoint inhibitors by inducing effector T-cell infiltration into the tumors and immune checkpoint signals. The combination of cancer vaccine and an immune checkpoint inhibitor may function synergistically to induce more effective antitumor immune responses, and clinical trials to test the combination are currently ongoing.
Collapse
Affiliation(s)
- Jennifer Kleponis
- 1 Department of Oncology, Department of Surgery, The Sidney Kimmel Comprehensive Cancer, The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA ; 2 Masters of Health Science Program in Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21287, USA
| | - Richard Skelton
- 1 Department of Oncology, Department of Surgery, The Sidney Kimmel Comprehensive Cancer, The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA ; 2 Masters of Health Science Program in Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21287, USA
| | - Lei Zheng
- 1 Department of Oncology, Department of Surgery, The Sidney Kimmel Comprehensive Cancer, The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA ; 2 Masters of Health Science Program in Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21287, USA
| |
Collapse
|
4133
|
Shirai T, Sano T, Kamijo F, Saito N, Miyake T, Kodaira M, Katoh N, Nishie K, Okuyama R, Uhara H. Acetylcholine receptor binding antibody-associated myasthenia gravis and rhabdomyolysis induced by nivolumab in a patient with melanoma. Jpn J Clin Oncol 2015; 46:86-8. [DOI: 10.1093/jjco/hyv158] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/26/2015] [Indexed: 01/03/2023] Open
|
4134
|
Teixidó C, González-Cao M, Karachaliou N, Rosell R. Predictive factors for immunotherapy in melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:208. [PMID: 26488004 DOI: 10.3978/j.issn.2305-5839.2015.05.07] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immunotherapy has emerged as an exciting strategy for cancer treatment. Therapeutic blockade of immune checkpoint regulators favors the ability of T cell responses to increase anti-tumor immunity. The cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death-1 (PD-1) are two T cell-inhibitory receptors with independent mechanisms of action. Immune checkpoint inhibitors targeting either CTLA-4, PD-1 or its ligand PD-L1 are currently yielding promising results in terms of efficacy in several clinical studies with melanoma patients and are being developed and tested as immunotherapy agents for multiple cancer types. To date, no reliable predictors of activity and efficacy of immunotherapy have yet been identified or validated. Even so, determining which patients derive clinical benefit from immune checkpoint agents remains an important clinical question and efforts to identify predictive markers of response are ongoing. This article reviews the current potential predictive factors for CTLA-4 and PD-1/PD-L1 immune checkpoints inhibitors in melanoma.
Collapse
Affiliation(s)
- Cristina Teixidó
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Maria González-Cao
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Niki Karachaliou
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Rafael Rosell
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| |
Collapse
|
4135
|
Abstract
Although head and neck squamous cell carcinoma has traditionally been considered to be a very immunosuppressive, or at least nonimmunogenic, tumor type, recent results from clinical studies of immune checkpoint blockade strategies have led to resurgence in the enthusiasm for immunotherapeutic approaches. Additional strategies for immunotherapy that are under active investigation include enhancement of cetuximab-mediated antibody-dependent cell-mediated cytotoxicity, tumor vaccines, and engineered T cells for adoptive therapy. All of these studies have early-phase clinical trials under way, and the next several years will be exciting as the results of these studies are reported.
Collapse
Affiliation(s)
- David W Schoppy
- Division of Head and Neck Surgery, Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John B Sunwoo
- Division of Head and Neck Surgery, Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
4136
|
Abstract
The development of immunotherapy using checkpoint blockade has altered the treatment landscape for patients who had but few options only several years ago. Currently, approved anti-checkpoint agents include ipilimumab, the first approved treatment aimed against the cytotoxic T-lymphocyte antigen-4 (CTLA-4) pathway, and pembrolizumab and nivolumab, which inhibit the programmed death-1 (PD-1) pathway. Careful monitoring and early intervention for immune-mediated side effects is important to mitigate toxicity. Immune-mediated response patterns may differ from response associated with conventional therapies, and so it is important to use caution against early abandonment of treatment. Biomarkers as predictive and prognostic markers of efficacy are still under investigation in an attempt to guide treatment selection in patients with advanced melanoma, and additional studies are needed to provide guidance for selection of checkpoint inhibitors to be used in sequence or combination.
Collapse
Affiliation(s)
- Sanjiv S Agarwala
- Chief of Medical Oncology and Hematology, St Luke's University Hospital and Health Network; Professor of Medicine Temple University, School of Medicine, Philadelphia, PA.
| |
Collapse
|
4137
|
Iacovelli R, Nolè F, Verri E, Renne G, Paglino C, Santoni M, Cossu Rocca M, Giglione P, Aurilio G, Cullurà D, Cascinu S, Porta C. Prognostic Role of PD-L1 Expression in Renal Cell Carcinoma. A Systematic Review and Meta-Analysis. Target Oncol 2015; 11:143-8. [DOI: 10.1007/s11523-015-0392-7] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
4138
|
Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM. Genetic Landscape and Biomarkers of Hepatocellular Carcinoma. Gastroenterology 2015; 149:1226-1239.e4. [PMID: 26099527 DOI: 10.1053/j.gastro.2015.05.061] [Citation(s) in RCA: 940] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/08/2015] [Accepted: 05/20/2015] [Indexed: 12/02/2022]
Abstract
Hepatocellular carcinoma (HCC) has emerged as a major cause of cancer-related death. Its mortality has increased in Western populations, with a minority of patients diagnosed at early stages, when curative treatments are feasible. Only the multikinase inhibitor sorafenib is available for the management of advanced cases. During the last 10 years, there has been a clear delineation of the landscape of genetic alterations in HCC, including high-level DNA amplifications in chromosome 6p21 (VEGFA) and 11q13 (FGF19/CNND1), as well as homozygous deletions in chromosome 9 (CDKN2A). The most frequent mutations affect TERT promoter (60%), associated with an increased telomerase expression. TERT promoter can also be affected by copy number variations and hepatitis B DNA insertions, and it can be found mutated in preneoplastic lesions. TP53 and CTNNB1 are the next most prevalent mutations, affecting 25%-30% of HCC patients, that, in addition to low-frequency mutated genes (eg, AXIN1, ARID2, ARID1A, TSC1/TSC2, RPS6KA3, KEAP1, MLL2), help define some of the core deregulated pathways in HCC. Conceptually, some of these changes behave as prototypic oncogenic addiction loops, being ideal biomarkers for specific therapeutic approaches. Data from genomic profiling enabled a proposal of HCC in 2 major molecular clusters (proliferation and nonproliferation), with differential enrichment in prognostic signatures, pathway activation and tumor phenotype. Translation of these discoveries into specific therapeutic decisions is an unmet medical need in this field.
Collapse
Affiliation(s)
- Jessica Zucman-Rossi
- Inserm, UMR-1162, Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue Contre le Cancer, Institut Universitaire d'Hematologie, Paris, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; Université Paris 13, Sorbonne Paris Cité, Unité de Formation et de Recherche Santé, Médecine, Biologie Humaine, Bobigny, France; Université Paris Diderot, Paris.
| | - Augusto Villanueva
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jean-Charles Nault
- Inserm, UMR-1162, Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue Contre le Cancer, Institut Universitaire d'Hematologie, Paris, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; Service d'hépatologie, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France
| | - Josep M Llovet
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Liver Cancer Translational Research Laboratory, Barcelona-Clínic Liver Cancer Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Liver Unit, CIBEREHD, Hospital Clínic, Barcelona, Catalonia, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
| |
Collapse
|
4139
|
Rogiers A, van den Oord JJ, Garmyn M, Stas M, Kenis C, Wildiers H, Marine JC, Wolter P. Novel Therapies for Metastatic Melanoma: An Update on Their Use in Older Patients. Drugs Aging 2015; 32:821-34. [PMID: 26442859 DOI: 10.1007/s40266-015-0304-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cutaneous melanoma is the most aggressive form of skin cancer. With age as a risk factor, melanoma is projected to become a substantial healthcare burden. The clinical course of melanoma in older patients is different from that in middle-aged and younger patients: melanomas are thicker, have higher mitotic rates and are more likely to be ulcerated. Older patients also have a higher mortality rate, yet, paradoxically, have a lower rate of lymph node metastases. After decades of no significant progress in the treatment of this devastating disease, novel insights into the mechanisms underlying the pathophysiology of metastatic melanoma have led to new and remarkably efficient therapeutic opportunities. The discovery that about half of all melanomas carry BRAF mutations led to the introduction of targeted therapy with significant improvements in clinical outcomes. Although these drugs appear to be equally effective in older patients, specific considerations regarding adverse events are required. Besides targeted therapy, immunotherapy has emerged as an alternative therapeutic option. Antibodies that block cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) can induce responses with high durability. Despite an aging immune system, older patients seem to benefit to the same degree from these treatments, apparently without increased toxicity. In this review, we focus on the epidemiology, clinicopathological features, and recent developments of systemic treatment in cutaneous melanoma with regard to older patients.
Collapse
Affiliation(s)
- Aljosja Rogiers
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Marjan Garmyn
- Department of Dermatology, University Hospitals Leuven, Leuven, Belgium
| | - Marguerite Stas
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Cindy Kenis
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Pascal Wolter
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
4140
|
Redman JM, Hill EM, AlDeghaither D, Weiner LM. Mechanisms of action of therapeutic antibodies for cancer. Mol Immunol 2015; 67:28-45. [PMID: 25911943 PMCID: PMC4529810 DOI: 10.1016/j.molimm.2015.04.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/29/2015] [Accepted: 04/03/2015] [Indexed: 02/06/2023]
Abstract
The therapeutic utility of antibodies and their derivatives is achieved by various means. The FDA has approved several targeted antibodies that disrupt signaling of various growth factor receptors for the treatment of a number of cancers. Rituximab, and other anti-CD20 monoclonal antibodies are active in B cell malignancies. As more experience has been gained with anti-CD20 monoclonal antibodies, the multifactorial nature of their anti-tumor mechanisms has emerged. Other targeted antibodies function to dampen inhibitory checkpoints. These checkpoint inhibitors have recently achieved dramatic results in several cancers, including melanoma. These and related antibodies continue to be investigated in the clinical and pre-clinical settings. Novel antibody structures that target two or more antigens have also made their way into clinical use. Tumor targeted antibodies can also be conjugated to chemo- or radiotherapeutic agents, or catalytic toxins, as a means to deliver toxic payloads to cancer cells. Here we provide a review of these mechanisms and a discussion of their relevance to current and future clinical applications.
Collapse
Affiliation(s)
- J M Redman
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - E M Hill
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - D AlDeghaither
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - L M Weiner
- Departments of Oncology and Internal Medicine, Georgetown University Medical Center and Lombardi Comprehensive Cancer Center, Washington, DC, United States.
| |
Collapse
|
4141
|
Karachaliou N, Pilotto S, Teixidó C, Viteri S, González-Cao M, Riso A, Morales-Espinosa D, Molina MA, Chaib I, Santarpia M, Richardet E, Bria E, Rosell R. Melanoma: oncogenic drivers and the immune system. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:265. [PMID: 26605311 PMCID: PMC4630557 DOI: 10.3978/j.issn.2305-5839.2015.08.06] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 12/19/2022]
Abstract
Advances and in-depth understanding of the biology of melanoma over the past 30 years have contributed to a change in the consideration of melanoma as one of the most therapy-resistant malignancies. The finding that oncogenic BRAF mutations drive tumor growth in up to 50% of melanomas led to a molecular therapy revolution for unresectable and metastatic disease. Moving beyond BRAF, inactivation of immune regulatory checkpoints that limit T cell responses to melanoma has provided targets for cancer immunotherapy. In this review, we discuss the molecular biology of melanoma and we focus on the recent advances of molecularly targeted and immunotherapeutic approaches.
Collapse
|
4142
|
PD-L1 expression as a potential predictive biomarker. Lancet Oncol 2015; 16:1285-7. [DOI: 10.1016/s1470-2045(15)00307-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 09/02/2015] [Accepted: 09/02/2015] [Indexed: 11/24/2022]
|
4143
|
|
4144
|
Socinski MA. Incorporating Immunotherapy Into the Treatment of Non-Small Cell Lung Cancer: Practical Guidance for the Clinic. Semin Oncol 2015; 42 Suppl 2:S19-28. [DOI: 10.1053/j.seminoncol.2015.09.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
4145
|
Tarhini AA, Zahoor H, Yearley JH, Gibson C, Rahman Z, Dubner R, Rao UNM, Sander C, Kirkwood JM. Tumor associated PD-L1 expression pattern in microscopically tumor positive sentinel lymph nodes in patients with melanoma. J Transl Med 2015; 13:319. [PMID: 26419843 PMCID: PMC4589168 DOI: 10.1186/s12967-015-0678-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 09/21/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Characterization of PD-L1 expression within clinically/radiologically negative but microscopically tumor positive sentinel lymph nodes (SLN) is important to our understanding of the relevance of this immune checkpoint pathway for adjuvant therapy. METHODS Patients included had primary cutaneous melanoma, Breslow thickness of 2.01-4.0 or >4 mm with or without tumor ulceration (T3a, T3b, T4a, T4b). All patients had microscopically tumor positive SLN. Hematoxylin and eosin (H&E) staining was performed, followed by PD-L1 immunohistochemical (IHC) staining using a preliminary IHC assay with anti-PD-L1 antibody clone 22C3. The slides were separately evaluated by two pathologists (JY and CG). Samples containing metastatic melanoma lesions were scored separately for PD-L1 expression in intratumoral and peritumoral locations, by utilizing two scoring methods. RESULTS Twenty-four patients where metastatic melanoma presence in the SLN was confirmed by H&E review of the cut sections were included in the final analysis of PD-L1 expression. SLN tumor size ranged from 1 to 2 mm. For three patients, the melanin content was too high to confidently assign a PD-L1 score. For the remaining 21 patients, all had some evidence of either intratumoral or peritumoral PD-L1 expression. The frequency of intratumoral tumor-associated PD-L1 expression was: 0 % of tumor cells (3 pts, 14 %); <1 % (5 pts, 24 %); 1-10 % (6 pts, 29 %) and >10 % (7 pts, 33 %). CONCLUSIONS Tumor-associated PD-L1 expression is readily detectable within melanoma micrometastases in the SLN of the majority of patients. These results support the testing of a therapeutic role for PD1/PD-L1 inhibition in the adjuvant setting, targeting melanoma micrometastases.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- University of Pittsburgh, Pittsburgh, PA, USA.
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.
| | - Haris Zahoor
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.
| | | | | | - Zahra Rahman
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.
| | - Rachel Dubner
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.
| | - Uma N M Rao
- University of Pittsburgh, Pittsburgh, PA, USA.
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.
| | | | - John M Kirkwood
- University of Pittsburgh, Pittsburgh, PA, USA.
- University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.
| |
Collapse
|
4146
|
Immunotherapy: Anti-PD-1 therapies-a new first-line option in advanced melanoma. Nat Rev Clin Oncol 2015; 12:625-6. [PMID: 26416151 DOI: 10.1038/nrclinonc.2015.170] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
4147
|
Eigentler TK. [Not Available]. J Dtsch Dermatol Ges 2015; 13:1055-6. [PMID: 26408485 DOI: 10.1111/ddg.12823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
4148
|
Chang GA, Tadepalli JS, Shao Y, Zhang Y, Weiss S, Robinson E, Spittle C, Furtado M, Shelton DN, Karlin-Neumann G, Pavlick A, Osman I, Polsky D. Sensitivity of plasma BRAFmutant and NRASmutant cell-free DNA assays to detect metastatic melanoma in patients with low RECIST scores and non-RECIST disease progression. Mol Oncol 2015; 10:157-65. [PMID: 26440707 DOI: 10.1016/j.molonc.2015.09.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/03/2015] [Indexed: 01/06/2023] Open
Abstract
Melanoma lacks a clinically useful blood-based biomarker of disease activity to help guide patient management. To determine whether measurements of circulating, cell-free, tumor-associated BRAF(mutant) and NRAS(mutant) DNA (ctDNA) have a higher sensitivity than LDH to detect metastatic disease prior to treatment initiation and upon disease progression we studied patients with unresectable stage IIIC/IV metastatic melanoma receiving treatment with BRAF inhibitor therapy or immune checkpoint blockade and at least 3 plasma samples obtained during their treatment course. Levels of BRAF(mutant) and NRAS(mutant) ctDNA were determined using droplet digital PCR (ddPCR) assays. Among patients with samples available prior to treatment initiation ctDNA and LDH levels were elevated in 12/15 (80%) and 6/20 (30%) (p = 0.006) patients respectively. In patients with RECIST scores <5 cm prior to treatment initiation, ctDNA levels were elevated in 5/7 (71%) patients compared to LDH which was elevated in 1/13 (8%) patients (p = 0.007). Among all disease progression events the modified bootstrapped sensitivities for ctDNA and LDH were 82% and 40% respectively, with a median difference in sensitivity of 42% (95% confidence interval, 27%-58%; P < 0.001). In addition, ctDNA levels were elevated in 13/16 (81%) instances of non-RECIST disease progression, including 10/12 (83%) instances of new brain metastases. In comparison LDH was elevated 8/16 (50%) instances of non-RECIST disease progression, including 6/12 (50%) instances of new brain metastases. Overall, ctDNA had a higher sensitivity than LDH to detect disease progression, including non-RECIST progression events. ctDNA has the potential to be a useful biomarker for monitoring melanoma disease activity.
Collapse
Affiliation(s)
- Gregory A Chang
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA
| | - Jyothirmayee S Tadepalli
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA
| | - Yongzhao Shao
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA; Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Yilong Zhang
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA; Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Sarah Weiss
- Department of Medicine, Division of Oncology, New York University School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA
| | - Eric Robinson
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA
| | | | - Manohar Furtado
- Digital Biology Center, Bio-Rad Laboratories, Pleasanton, CA, USA
| | - Dawne N Shelton
- Digital Biology Center, Bio-Rad Laboratories, Pleasanton, CA, USA
| | | | - Anna Pavlick
- Department of Medicine, Division of Oncology, New York University School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA
| | - Iman Osman
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA
| | - David Polsky
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, USA.
| |
Collapse
|
4149
|
Bui JK, Mellors JW. Reversal of T-cell exhaustion as a strategy to improve immune control of HIV-1. AIDS 2015; 29:1911-5. [PMID: 26355569 DOI: 10.1097/qad.0000000000000788] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
4150
|
Zheng P, Zhou Z. Human Cancer Immunotherapy with PD-1/PD-L1 Blockade. BIOMARKERS IN CANCER 2015; 7:15-8. [PMID: 26448693 PMCID: PMC4578571 DOI: 10.4137/bic.s29325] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/19/2015] [Accepted: 07/22/2015] [Indexed: 01/06/2023]
Abstract
The ligation of programmed cell death-1 (PD-1) to its ligands PD-L1 and PD-L2 counteracts T-cell activation, which is critical in immune tolerance. The persistent high expression of PD-1 and PD-L1 are also observed on tumor-infiltrating lymphocytes and various tumor cells, maintaining the highly suppressive microenvironment in tumor sites and promoting tumor malignancies. The blockade of PD-1 axis with PD-L2 fusion protein or monoclonal antibodies against either PD-1 or PD-L1 has been clinically evaluated in various tumor types. This short review summarizes the progress of PD-1 axis blockade in clinical trials to evaluate its effectiveness in the antitumor immunotherapy.
Collapse
Affiliation(s)
- Peilin Zheng
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China
| | - Zhiguang Zhou
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China
| |
Collapse
|