The Changing Landscape of Immunotherapy for Advanced Renal Cancer

Natural history and therapy of metastatic renal cell carcinoma: the role of interleukin-2.

Cancer. 80: 1198-1220

Interferon-alpha and survival in metastatic renal carcinoma: early results of a randomised controlled trial. Medical Research Council Renal Cancer Collaborators.

Lancet. 353: 14-17Motzer R.J. Hutson T.E. Tomczak P. et al.

Sunitinib versus interferon alfa in metastatic renal-cell carcinoma.

N Engl J Med. 356: 115-124Hudes G. Carducci M. Tomczak P. et al.

Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma.

N Engl J Med. 356: 2271-2281Motzer R.J. Escudier B. Oudard S. et al.

Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial.

Lancet. 372: 449-456Sternberg C.N. Davis I.D. Mardiak J. et al.

Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial.

J Clin Oncol. 28: 1061-1068Rini B.I. Escudier B. Tomczak P. et al.

Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial.

Lancet. 378: 1931-1939Postow M.A. Callahan M.K. Wolchok J.D.

Immune checkpoint blockade in cancer therapy.

J Clin Oncol. 33: 1974-1982Motzer R.J. Escudier B. McDermott D.F. et al.

Nivolumab versus everolimus in advanced renal-cell carcinoma.

N Engl J Med. 373: 1803-1813Motzer R.J. Jonasch E. Agarwal N. et al.

Kidney cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology.

J Natl Compr Canc Netw. 20: 71-90

Spontaneous regression of cancer and the importance of finding its cause.

Natl Cancer Inst Monogr. 44: 5-9Motzer R.J. Bander N.H. Nanus D.M.

Renal-cell carcinoma.

N Engl J Med. 335: 865-875Klapper J.A. Downey S.G. Smith F.O. et al.

High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma: a retrospective analysis of response and survival in patients treated in the surgery branch at the National Cancer Institute between 1986 and 2006.

Cancer. 113: 293-301Rosenberg S.A. Lotze M.T. Muul L.M. et al.

A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin-2 alone.

N Engl J Med. 316: 889-897Fisher R.I. Coltman Jr., C.A. Doroshow J.H. et al.

Metastatic renal cancer treated with interleukin-2 and lymphokine-activated killer cells. A phase II clinical trial.

Ann Intern Med. 108: 518-523Negrier S. Philip T. Stoter G. et al.

Interleukin-2 with or without LAK cells in metastatic renal cell carcinoma: a report of a European multicentre study.

Eur J Cancer Clin Oncol. 25: S21-S28Childs R.W. Clave E. Tisdale J. et al.

Successful treatment of metastatic renal cell carcinoma with a nonmyeloablative allogeneic peripheral-blood progenitor-cell transplant: evidence for a graft-versus-tumor effect.

J Clin Oncol. 17: 2044-2049

Nonmyeloablative allogeneic peripheral blood stem-cell transplantation as immunotherapy for malignant diseases.

Cancer J. 6: 179-187

Self-tolerance: context dependent tuning of T cell antigen recognition.

Semin Immunol. 12 (): 197-203Goodnow C.C. Sprent J. Fazekas de St Groth B. et al.

Cellular and genetic mechanisms of self tolerance and autoimmunity.

Nature. 435: 590-597

Regulatory T cells: key controllers of immunologic self-tolerance.

Cell. 101: 455-458

Molecular and cellular insights into T cell exhaustion.

Nat Rev Immunol. 15: 486-499Lafferty K.J. Misko I.S. Cooley M.A.

Allogeneic stimulation modulates the in vitro response of T cells to transplantation antigen.

Nature. 249: 275-276Riley J.L. Mao M. Kobayashi S. et al.

Modulation of TCR-induced transcriptional profiles by ligation of CD28, ICOS, and CTLA-4 receptors.

Proc Natl Acad Sci U S A. 99: 11790-11795Diehn M. Alizadeh A.A. Rando O.J. et al.

Genomic expression programs and the integration of the CD28 costimulatory signal in T cell activation.

Proc Natl Acad Sci U S A. 99: 11796-11801Brunet J.F. Denizot F. Luciani M.F. et al.

A new member of the immunoglobulin superfamily--CTLA-4.

Nature. 328: 267-270Dariavach P. Mattéi M.G. Golstein P. et al.

Human Ig superfamily CTLA-4 gene: chromosomal localization and identity of protein sequence between murine and human CTLA-4 cytoplasmic domains.

Eur J Immunol. 18: 1901-1905Linsley P.S. Greene J.L. Brady W. et al.

Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors.

Immunity. 1: 793-801Parry R.V. Chemnitz J.M. Frauwirth K.A. et al.

CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms.

Mol Cell Biol. 25: 9543-9553Freeman G.J. Long A.J. Iwai Y. et al.

Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation.

J Exp Med. 192: 1027-1034Iwai Y. Ishida M. Tanaka Y. et al.

Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade.

Proc Natl Acad Sci U S A. 99: 12293-12297Hui E. Cheung J. Zhu J. et al.

T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition.

Science. 355: 1428-1433Leach D.R. Krummel M.F. Allison J.P.

Enhancement of antitumor immunity by CTLA-4 blockade.

Science. 271: 1734-1736Waldman A.D. Fritz J.M. Lenardo M.J.

A guide to cancer immunotherapy: from T cell basic science to clinical practice.

Nat Rev Immunol. 20: 651-668Krummel M.F. Allison J.P.

CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation.

J Exp Med. 182: 459-465Tivol E.A. Borriello F. Schweitzer A.N. et al.

Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4.

Immunity. 3: 541-547Hodi F.S. O'Day S.J. McDermott D.F. et al.

Improved survival with ipilimumab in patients with metastatic melanoma.

N Engl J Med. 363: 711-723Ishida Y. Agata Y. Shibahara K. et al.

Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death.

Embo J. 11: 3887-3895Nishimura H. Nose M. Hiai H. et al.

Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor.

Immunity. 11: 141-151Iwai Y. Terawaki S. Honjo T.

PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells.

Int Immunol. 17: 133-144Hirano F. Kaneko K. Tamura H. et al.

Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity.

Cancer Res. 65: 1089-1096Barber D.L. Wherry E.J. Masopust D. et al.

Restoring function in exhausted CD8 T cells during chronic viral infection.

Nature. 439: 682-687Topalian S.L. Hodi F.S. Brahmer J.R. et al.

Safety, activity, and immune correlates of anti-PD-1 antibody in cancer.

N Engl J Med. 366: 2443-2454Michot J.M. Bigenwald C. Champiat S. et al.

Immune-related adverse events with immune checkpoint blockade: a comprehensive review.

Eur J Cancer. 54: 139-148Motzer R.J. Escudier B. George S. et al.

Nivolumab versus everolimus in patients with advanced renal cell carcinoma: updated results with long-term follow-up of the randomized, open-label, phase 3 CheckMate 025 trial.

Cancer. 126: 4156-4167Motzer R.J. Tannir N.M. McDermott D.F. et al.

Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma.

N Engl J Med. 378: 1277-1290Motzer R.J. McDermott D.F. Escudier B. et al.

Conditional survival and long-term efficacy with nivolumab plus ipilimumab versus sunitinib in patients with advanced renal cell carcinoma.

Cancer. 128: 2085-2097Brahmer J.R. Lacchetti C. Schneider B.J. et al.

Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline.

J Clin Oncol. 36: 1714-1768

Von Hippel-Lindau disease.

Annu Rev Pathol. 2: 145-173Sgambati M.T. Stolle C. Choyke P.L. et al.

Mosaicism in von Hippel-Lindau disease: lessons from kindreds with germline mutations identified in offspring with mosaic parents.

Am J Hum Genet. 66: 84-91Latif F. Tory K. Gnarra J. et al.

Identification of the von Hippel-Lindau disease tumor suppressor gene.

Science. 260: 1317-1320

Comprehensive molecular characterization of clear cell renal cell carcinoma.

Nature. 499: 43-49Bratslavsky G. Sudarshan S. Neckers L. et al.

Pseudohypoxic pathways in renal cell carcinoma.

Clin Cancer Res. 13: 4667-4671

The von Hippel-Lindau tumour suppressor protein: O2 sensing and cancer.

Nat Rev Cancer. 8: 865-873Choueiri T.K. Kaelin Jr., W.G.

Targeting the HIF2-VEGF axis in renal cell carcinoma.

Nat Med. 26: 1519-1530https://doi.org/10.1038/s41591-020-1093-zSaxton R.A. Sabatini D.M.

mTOR signaling in growth, metabolism, and disease.

Cell. 168: 960-976Toschi A. Lee E. Gadir N. et al.

Differential dependence of hypoxia-inducible factors 1 alpha and 2 alpha on mTORC1 and mTORC2.

J Biol Chem. 283: 34495-34499van Beijnum J.R. Nowak-Sliwinska P. Huijbers E.J. et al.

The great escape; the hallmarks of resistance to antiangiogenic therapy.

Pharmacol Rev. 67: 441-461Gabrilovich D.I. Ciernik I.F. Carbone D.P.

Dendritic cells in antitumor immune responses. I. Defective antigen presentation in tumor-bearing hosts.

Cell Immunol. 170: 101-110Chaux P. Favre N. Martin M. et al.

Tumor-infiltrating dendritic cells are defective in their antigen-presenting function and inducible B7 expression in rats.

Int J Cancer. 72: 619-624Gabrilovich D.I. Corak J. Ciernik I.F. et al.

Decreased antigen presentation by dendritic cells in patients with breast cancer.

Clin Cancer Res. 3: 483-490Gabrilovich D. Ishida T. Oyama T. et al.

Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo.

Blood. 92: 4150-4166Hegde P.S. Wallin J.J. Mancao C.

Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics.

Semin Cancer Biol. 52: 117-124Fukumura D. Kloepper J. Amoozgar Z. et al.

Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges.

Nat Rev Clin Oncol. 15: 325-340Gabrilovich D.I. Chen H.L. Girgis K.R. et al.

Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells.

Nat Med. 2: 1096-1103Wallin J.J. Bendell J.C. Funke R. et al.

Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma.

Nat Commun. 7: 12624

Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy.

Nat Med. 7: 987-989Yuan H. Cai P. Li Q. et al.

Axitinib augments antitumor activity in renal cell carcinoma via STAT3-dependent reversal of myeloid-derived suppressor cell accumulation.

Biomed Pharmacother. 68: 751-756Rini B.I. Plimack E.R. Stus V. et al.

Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma.

N Engl J Med. 380: 1116-1127Powles T. Plimack E.R. Soulières D. et al.

Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE–426): extended follow-up from a randomised, open-label, phase 3 trial.

Lancet Oncol. 21: 1563-1573Motzer R.J. Penkov K. Haanen J. et al.

Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma.

N Engl J Med. 380: 1103-1115Choueiri T.K. Motzer R.J. Rini B.I. et al.

Updated efficacy results from the JAVELIN Renal 101 trial: first-line avelumab plus axitinib versus sunitinib in patients with advanced renal cell carcinoma.

Ann Oncol. 31: 1030-1039Motzer R. Alekseev B. Rha S.Y. et al.

Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma.

N Engl J Med. 384: 1289-1300Choueiri T.K. Powles T. Burotto M. et al.

Nivolumab plus cabozantinib versus sunitinib for advanced renal-cell carcinoma.

N Engl J Med. 384: 829-841Motzer R.J. Powles T. Burotto M. et al.

Nivolumab plus cabozantinib versus sunitinib in first-line treatment for advanced renal cell carcinoma (CheckMate 9ER): long-term follow-up results from an open-label, randomised, phase 3 trial.

Lancet Oncol. 23: 888-898Rini B.I. Powles T. Atkins M.B. et al.

Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma (IMmotion151): a multicentre, open-label, phase 3, randomised controlled trial.

Lancet. 393: 2404-2415Motzer R.J. Powles T. Atkins M.B. et al.

Final overall survival and molecular analysis in IMmotion151, a phase 3 trial comparing atezolizumab plus bevacizumab vs sunitinib in patients with previously untreated metastatic renal cell carcinoma.

JAMA Oncol. 8: 275-280Hahn A.W. Msaouel P. Tannir N.M.

CLEAR Trial: is lenvatinib plus pembrolizumab the best first-line immunotherapy doublet in metastatic renal cell carcinoma? In: The ASCO Post.

() ()Cella D. Motzer R.J. Suarez C. et al.

Patient-reported outcomes with first-line nivolumab plus cabozantinib versus sunitinib in patients with advanced renal cell carcinoma treated in CheckMate 9ER: an open-label, randomised, phase 3 trial.

Lancet Oncol. 23: 292-303McGregor B.A. McKay R.R. Braun D.A. et al.

Results of a multicenter phase II study of atezolizumab and bevacizumab for patients with metastatic renal cell carcinoma with variant histology and/or sarcomatoid features.

J Clin Oncol. 38: 63-70Lee C.H. Voss M.H. Carlo M.I. et al.

Phase II trial of cabozantinib plus nivolumab in patients with non-clear-cell renal cell carcinoma and genomic correlates.

J Clin Oncol. 40: 2333-2341Lee C.-H. Li C. Perini R.F. et al.

KEYNOTE-B61: open-label phase 2 study of pembrolizumab in combination with lenvatinib as first-line treatment for non-clear cell renal cell carcinoma (nccRCC).

J Clin Oncol. 39Haas N.B. Manola J. Uzzo R.G. et al.

Adjuvant sunitinib or sorafenib for high-risk, non-metastatic renal-cell carcinoma (ECOG-ACRIN E2805): a double-blind, placebo-controlled, randomised, phase 3 trial.

Lancet. 387: 2008-2016Haas N.B. Manola J. Dutcher J.P. et al.

Adjuvant treatment for high-risk clear cell renal cancer: updated results of a high-risk subset of the ASSURE randomized trial.

JAMA Oncol. 3: 1249-1252Motzer R.J. Haas N.B. Donskov F. et al.

Randomized phase III trial of adjuvant pazopanib versus placebo after nephrectomy in patients with localized or locally advanced renal cell carcinoma.

J Clin Oncol. 35: 3916-3923Sun M. Marconi L. Eisen T. et al.

Adjuvant vascular endothelial growth factor-targeted therapy in renal cell carcinoma: a systematic review and pooled analysis.

Eur Urol. Nov. 74: 611-620Gross-Goupil M. Kwon T.G. Eto M. et al.

Axitinib versus placebo as an adjuvant treatment of renal cell carcinoma: results from the phase III, randomized ATLAS trial.

Ann Oncol. 29: 2371-2378Ravaud A. Motzer R.J. Pandha H.S. et al.

Adjuvant sunitinib in high-risk renal-cell carcinoma after nephrectomy.

N Engl J Med. 375: 2246-2254https://doi.org/10.1056/NEJMoa1611406Choueiri T.K. Tomczak P. Park S.H. et al.

Adjuvant pembrolizumab after nephrectomy in renal-cell carcinoma.

N Engl J Med. 385: 683-694Powles T. Tomczak P. Park S.H. et al.

Pembrolizumab versus placebo as post-nephrectomy adjuvant therapy for clear cell renal cell carcinoma (KEYNOTE–564): 30-month follow-up analysis of a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial.

Lancet Oncol. 23: 1133-1144Bex A. Russo P. Tomita Y. et al.

A phase III, randomized, placebo-controlled trial of nivolumab or nivolumab plus ipilimumab in patients with localized renal cell carcinoma at high-risk of relapse after radical or partial nephrectomy (CheckMate 914).

American Society of Clinical Oncology. 38: supplPal S.K. Uzzo R. Karam J.A. et al.

Adjuvant atezolizumab versus placebo for patients with renal cell carcinoma at increased risk of recurrence following resection (IMmotion010): a multicentre, randomised, double-blind, phase 3 trial.

Lancet. https://doi.org/10.1016/s0140-6736(22)01658-0Haas N.B. Puligandla M. Allaf M.E. et al.

PROSPER: phase III randomized study comparing perioperative nivolumab versus observation in patients with renal cell carcinoma (RCC) undergoing nephrectomy (ECOG-ACRIN EA8143).

American Society of Clinical Oncology. 38: suppl

ESMO 2022: Phase 3 Study of Cabozantinib in Combination With Nivolumab and Ipilimumab in Previously Untreated Advanced Renal Cell Carcinoma of Imdc Intermediate or Poor Risk (COSMIC-313). In: ESMO 2022 Kidney Cancer.

() ()

ESMO 2022: Invited Discussant: Results of COSMIC-313. In: ESMO 2022 Kidney Cancer.

() ()Cristescu R. Mogg R. Ayers M. et al.

Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy.

Science. 362https://doi.org/10.1126/science.aar3593Le D.T. Uram J.N. Wang H. et al.

PD-1 blockade in tumors with mismatch-repair deficiency.

N Engl J Med. 372: 2509-2520Le D.T. Durham J.N. Smith K.N. et al.

Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade.

Science. 357: 409-413Bruni D. Angell H.K. Galon J.

The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy.

Nat Rev Cancer. 20: 662-680Braun D.A. Hou Y. Bakouny Z. et al.

Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma.

Nat Med. 26: 909-918Yarchoan M. Hopkins A. Jaffee E.M.

Tumor mutational burden and response rate to PD-1 inhibition.

N Engl J Med. 377: 2500-2501Latham A. Srinivasan P. Kemel Y. et al.

Microsatellite instability is associated with the presence of Lynch syndrome pan-cancer.

J Clin Oncol. 37: 286-295Mandal R. Samstein R.M. Lee K.W. et al.

Genetic diversity of tumors with mismatch repair deficiency influences anti-PD-1 immunotherapy response.

Science. 364: 485-491Motzer R.J. Choueiri T.K. McDermott D.F. et al.

Biomarker analysis from CheckMate 214: nivolumab plus ipilimumab versus sunitinib in renal cell carcinoma. J Immunother Cancer.

Mar. 10Braun D.A. Bakouny Z. Hirsch L. et al.

Beyond conventional immune-checkpoint inhibition: novel immunotherapies for renal cell carcinoma.

Nat Rev Clin Oncol. 18: 199-214Bakouny Z. Braun D.A. Shukla S.A. et al.

Integrative molecular characterization of sarcomatoid and rhabdoid renal cell carcinoma.

Nat Commun. 12: 808Tannir N.M. Signoretti S. Choueiri T.K. et al.

Efficacy and safety of nivolumab plus ipilimumab versus sunitinib in first-line treatment of patients with advanced sarcomatoid renal cell carcinoma.

Clin Cancer Res. 27: 78-86Kawakami F. Sircar K. Rodriguez-Canales J. et al.

Programmed cell death ligand 1 and tumor-infiltrating lymphocyte status in patients with renal cell carcinoma and sarcomatoid dedifferentiation.

Cancer. 123: 4823-4831Braun D.A. Street K. Burke K.P. et al.

Progressive immune dysfunction with advancing disease stage in renal cell carcinoma.

Cancer Cell. 39: 632-648.e8Kraehenbuehl L. Weng C.H. Eghbali S. et al.

Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways.

Nat Rev Clin Oncol. 19: 37-50Xu L. Zhu Y. Chen L. et al.

Prognostic value of diametrically polarized tumor-associated macrophages in renal cell carcinoma.

Ann Surg Oncol. 21: 3142-3150Hakimi A.A. Voss M.H. Kuo F. et al.

Transcriptomic profiling of the tumor microenvironment reveals distinct subgroups of clear cell renal cell cancer: data from a randomized phase III trial.

Cancer Discov. 9: 510-525Davidsson S. Fiorentino M. Giunchi F. et al.

Infiltration of M2 macrophages and regulatory T cells plays a role in recurrence of renal cell carcinoma.

Eur Urol Open. 20: 62-71Yuen K.C. Liu L.F. Gupta V. et al.

High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade.

Nat Med. 26: 693-698

IL-8 and cancer prognosis on immunotherapy.

Nat Med. 26: 650-651Sauer N. Szlasa W. Jonderko L. et al.

LAG-3 as a potent target for novel anticancer therapies of a wide range of tumors.

Int J Mol Sci. 23https://doi.org/10.3390/ijms23179958Florou V. Garrido-Laguna I.

Clinical development of anti-TIGIT antibodies for immunotherapy of cancer.

Curr Oncol Rep. 24: 1107-1112Annese T. Tamma R. Ribatti D.

Update in TIGIT immune-checkpoint role in cancer.

Front Oncol. 12: 871085Gomes de Morais A.L. Cerdá S. de Miguel M.

New checkpoint inhibitors on the road: targeting TIM-3 in solid tumors.

Curr Oncol Rep. 24: 651-658Hosono M. Koma Y.I. Takase N. et al.

CXCL8 derived from tumor-associated macrophages and esophageal squamous cell carcinomas contributes to tumor progression by promoting migration and invasion of cancer cells.

Oncotarget. 8: 106071-106088Korbecki J. Kupnicka P. Chlubek M. et al.

CXCR2 receptor: regulation of expression, signal transduction, and involvement in cancer.

Int J Mol Sci. 23https://doi.org/10.3390/ijms23042168Najjar Y.G. Rayman P. Jia X. et al.

Myeloid-derived suppressor cell subset accumulation in renal cell carcinoma parenchyma is associated with intratumoral expression of IL1β, IL8, CXCL5, and Mip-1α.

Clin Cancer Res. 23: 2346-2355Jaillon S. Ponzetta A. Di Mitri D. et al.

Neutrophil diversity and plasticity in tumour progression and therapy.

Nat Rev Cancer. 20: 485-503Uemura H. Fujimoto K. Tanaka M. et al.

A phase I trial of vaccination of CA9-derived peptides for HLA-A24-positive patients with cytokine-refractory metastatic renal cell carcinoma.

Clin Cancer Res. 12: 1768-1775

留言 (0)

沒有登入
gif