Transplant Onconephrology in Patients With Kidney Transplants

Blosser C.D. Haber G. Engels E.A.

Changes in cancer incidence and outcomes among kidney transplant recipients in the United States over a thirty-year period.

Kidney Int. 99: 1430-1438Al-Adra D.P. Hammel L. Roberts J. et al.

Pretransplant solid organ malignancy and organ transplant candidacy: a consensus expert opinion statement.

Am J Transpl. 21: 460-474Al-Adra D.P. Hammel L. Roberts J. et al.

Preexisting melanoma and hematological malignancies, prognosis, and timing to solid organ transplantation: a consensus expert opinion statement.

Am J Transpl. 21: 475-483Engels E.A. Pfeiffer R.M. Fraumeni J.F. et al.

Spectrum of cancer risk among US solid organ transplant recipients.

JAMA. 306: 1891-1901Acuna S.A. Huang J.W. Scott A.L. et al.

Cancer screening recommendations for solid organ transplant recipients: a systematic review of clinical practice guidelines.

Am J Transpl. 17: 103-114Kasiske B.L. Vazquez M.A. Harmon W.E. et al.

Recommendations for the outpatient surveillance of renal transplant recipients. American Society of Transplantation.

J Am Soc Nephrol. 11: S1-S86Doublet J.D. Peraldi M.N. Gattegno B. Thibault P. Sraer J.D.

Renal cell carcinoma of native kidneys: prospective study of 129 renal transplant patients.

J Urol. 158: 42-44Schwarz A. Vatandaslar S. Merkel S. Haller H.

Renal cell carcinoma in transplant recipients with acquired cystic kidney disease.

Clin J Am Soc Nephrol. 2: 750-756Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group

KDIGO clinical practice guideline for the care of kidney transplant recipients.

Am J Transpl. 9: S1-S155Mihalov M.L. Gattuso P. Abraham K. Holmes E.W. Reddy V.

Incidence of post-transplant malignancy among 674 solid-organ-transplant recipients at a single center.

Clin Transpl. 10: 248-255

Incidence of non-Hodgkin lymphoma in kidney and heart transplant recipients.

Lancet. 342: 1514-1516Cherikh W.S. Kauffman H.M. McBride M.A. Maghirang J. Swinnen L.J. Hanto D.W.

Association of the type of induction immunosuppression with posttransplant lymphoproliferative disorder, graft survival, and patient survival after primary kidney transplantation.

Transplantation. 76: 1289-1293Bustami R.T. Ojo A.O. Wolfe R.A. et al.

Immunosuppression and the risk of post-transplant malignancy among cadaveric first kidney transplant recipients.

Am J Transpl. 4: 87-93Marks W.H. Ilsley J.N. Dharnidharka V.R.

Posttransplantation lymphoproliferative disorder in kidney and heart transplant recipients receiving thymoglobulin: a systematic review.

Transpl Proc. 43: 1395-1404Herman M. Weinstein T. Korzets A. et al.

Effect of cyclosporin A on DNA repair and cancer incidence in kidney transplant recipients.

J Lab Clin Med. 137: 14-20Dantal J. Hourmant M. Cantarovich D. et al.

Effect of long-term immunosuppression in kidney-graft recipients on cancer incidence: randomised comparison of two cyclosporin regimens.

Lancet. 351: 623-628Dharnidharka V.R. Ho P.-L. Stablein D.M. Harmon W.E. Tejani A.H.

Mycophenolate, tacrolimus and post-transplant lymphoproliferative disorder: a report of the North American pediatric renal transplant Cooperative study.

Pediatr Transpl. 6: 396-399Swann P.F. Waters T.R. Moulton D.C. et al.

Role of postreplicative DNA mismatch repair in the cytotoxic action of thioguanine.

Science. 273: 1109-1111Offman J. Opelz G. Doehler B. et al.

Defective DNA mismatch repair in acute myeloid leukemia/myelodysplastic syndrome after organ transplantation.

Blood. 104: 822-828Robson R. Cecka J.M. Opelz G. Budde M. Sacks S.

Prospective registry-based observational cohort study of the long-term risk of malignancies in renal transplant patients treated with mycophenolate mofetil.

Am J Transpl. 5: 2954-2960Campistol J.M. Eris J. Oberbauer R. et al.

Sirolimus therapy after early cyclosporine withdrawal reduces the risk for cancer in adult renal transplantation.

J Am Soc Nephrol. 17: 581-589Luan F.L. Hojo M. Maluccio M. Yamaji K. Suthanthiran M.

Rapamycin blocks tumor progression: unlinking immunosuppression from antitumor efficacy.

Transplantation. 73: 1565-1572Stallone G. Schena A. Infante B. et al.

Sirolimus for Kaposi’s sarcoma in renal-transplant recipients.

N Engl J Med. 352: 1317-1323Euvrard S. Morelon E. Rostaing L. et al.

Sirolimus and secondary skin-cancer prevention in kidney transplantation.

N Engl J Med. 367: 329-339Santos A.H. Chen C. Leghrouz M.A. Bueno E.P. Lee J.J. Wen X.

Association of HLA mismatch and MTOR inhibitor regimens with malignancy and mortality after kidney transplantation.

Transpl Immunol. 66: 101391Muthukkumar S. Ramesh T.M. Bondada S.

Rapamycin, a potent immunosuppressive drug, causes programmed cell death in B lymphoma cells.

Transplantation. 60: 264-270Luan F.L. Ding R. Sharma V.K. Chon W.J. Lagman M. Suthanthiran M.

Rapamycin is an effective inhibitor of human renal cancer metastasis.

Kidney Int. 63: 917-926Nepomuceno R.R. Balatoni C.E. Natkunam Y. Snow A.L. Krams S.M. Martinez O.M.

Rapamycin inhibits the interleukin 10 signal transduction pathway and the growth of Epstein Barr virus B-cell lymphomas.

Cancer Res. 63: 4472-4480García-Morales P. Hernando E. Carrasco-García E. Menéndez-Gutierrez M.P. Saceda M. Martínez-Lacaci I.

Cyclin D3 is down-regulated by rapamycin in HER-2-overexpressing breast cancer cells.

Mol Cancer Ther. 5: 2172-2181Guba M. von Breitenbuch P. Steinbauer M. et al.

Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor.

Nat Med. 8: 128-135Huber S. Bruns C.J. Schmid G. et al.

Inhibition of the mammalian target of rapamycin impedes lymphangiogenesis.

Kidney Int. 71: 771-777Vincenti F. Charpentier B. Vanrenterghem Y. et al.

A phase III study of belatacept-based immunosuppression regimens versus cyclosporine in renal transplant recipients (BENEFIT study).

Am J Transpl. 10: 535-546Vincenti F. Rostaing L. Grinyo J. et al.

Belatacept and long-term outcomes in kidney transplantation.

N Engl J Med. 374: 333-343Dharnidharka V.R. Webster A.C. Martinez O.M. Preiksaitis J.K. Leblond V. Choquet S.

Post-transplant lymphoproliferative disorders.

Nat Rev Dis Primers. 2: 15088Allen U.D. Preiksaitis J.K. AST Infectious Diseases Community of Practice

Post-transplant lymphoproliferative disorders, Epstein-barr virus infection, and disease in solid organ transplantation: guidelines from the American Society of transplantation infectious diseases Community of practice.

Clin Transpl. 33: e13652Reshef R. Vardhanabhuti S. Luskin M.R. et al.

Reduction of immunosuppression as initial therapy for posttransplantation lymphoproliferative disorder(★).

Am J Transpl. 11: 336-347Trappe R.U. Dierickx D. Zimmermann H. et al.

Response to rituximab induction is a Predictive marker in B-cell post-transplant lymphoproliferative disorder and Allows successful stratification into rituximab or R-CHOP Consolidation in an International, prospective, multicenter phase II trial.

J Clin Oncol. 35: 536-543Zimmermann H. Trappe R.U.

EBV and posttransplantation lymphoproliferative disease: what to do?.

Hematol Am Soc Hematol Educ Program. 2013: 95-102Haque T. Wilkie G.M. Jones M.M. et al.

Allogeneic cytotoxic T-cell therapy for EBV-positive posttransplantation lymphoproliferative disease: results of a phase 2 multicenter clinical trial.

Blood. 110: 1123-1131Chiou F.K. Beath S.V. Wilkie G.M. Vickers M.A. Morland B. Gupte G.L.

Cytotoxic T-lymphocyte therapy for post-transplant lymphoproliferative disorder after solid organ transplantation in children.

Pediatr Transpl. 22https://doi.org/10.1111/petr.13133Neelapu S.S. Locke F.L. Bartlett N.L. et al.

Axicabtagene Ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma.

New Engl J Med. 377: 2531-2544Raje N. Berdeja J. Lin Y. et al.

Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma.

N Engl J Med. 380: 1726-1737Kanduri S.R. Cheungpasitporn W. Thongprayoon C. et al.

Systematic review of risk factors and incidence of acute kidney injury among patients treated with CAR-T cell therapies.

Kidney Int Rep. 6: 1416-1422Gupta S. Seethapathy H. Strohbehn I.A. et al.

Acute kidney injury and electrolyte abnormalities after chimeric antigen receptor T-cell (CAR-T) therapy for diffuse large B-cell lymphoma.

Am J Kidney Dis. 76: 63-71Le R.Q. Li L. Yuan W. et al.

FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome.

Oncologist. 23: 943-947Krishnamoorthy S. Ghobadi A. Santos R.D. et al.

CAR-T therapy in solid organ transplant recipients with treatment refractory posttransplant lymphoproliferative disorder.

Am J Transpl. 21: 809-814Mamlouk O. Nair R. Iyer S.P. et al.

Safety and efficacy of CAR T-cell therapy in kidney transplant recipients.

Blood. Luttwak E. Hagin D. Perry C. et al.

Anti-CD19 CAR-T therapy for EBV-negative posttransplantation lymphoproliferative disease-a single center case series.

Bone Marrow Transpl. 56: 1031-1037Decourt A. Gondouin B. Delaroziere J.C. et al.

Trends in survival and renal Recovery in patients with multiple myeloma or light-chain amyloidosis on Chronic dialysis.

Clin J Am Soc Nephrol. 11: 431-441Leung N. Lager D.J. Gertz M.A. Wilson K. Kanakiriya S. Fervenza F.C.

Long-term outcome of renal transplantation in light-chain deposition disease.

Am J Kidney Dis. 43: 147-153Kumar S.K. Dispenzieri A. Lacy M.Q. et al.

Continued improvement in survival in multiple myeloma: changes in early mortality and outcomes in older patients.

Leukemia. 28: 1122-1128Fonseca R. Abouzaid S. Bonafede M. et al.

Trends in overall survival and costs of multiple myeloma, 2000-2014.

Leukemia. 31: 1915-1921

Multiple myeloma current treatment algorithms.

Blood Cancer J. 10: 94Voorhees P.M. Kaufman J.L. Laubach J. et al.

Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial.

Blood. 136: 936-945Palumbo A. Avet-Loiseau H. Oliva S. et al.

Revised International staging system for multiple myeloma: a report from International myeloma working group.

J Clin Oncol. 33: 2863-2869Munshi N.C. Avet-Loiseau H. Rawstron A.C. et al.

Association of minimal residual disease with Superior survival outcomes in patients with multiple myeloma: a meta-analysis.

JAMA Oncol. 3: 28-35Chitty D.W. Hartley-Brown M.A. Abate M. et al.

Kidney transplantation in patients with multiple myeloma: narrative analysis and review of the last 2 decades.

Nephrol Dial Transpl. gfaa361https://doi.org/10.1093/ndt/gfaa361Pasternack A. Ahonen J. Kuhlbäck B.

Renal transplantation in 45 patients with amyloidosis.

Transplantation. 42: 598-601Angel-Korman A. Stern L. Sarosiek S. et al.

Long-term outcome of kidney transplantation in AL amyloidosis.

Kidney Int. 95: 405-411Law S. Cohen O. Lachmann H.J. et al.

Renal transplant outcomes in amyloidosis.

Nephrol Dial Transpl. 36: 355-365Kumar S.K. Therneau T.M. Gertz M.A. et al.

Clinical course of patients with relapsed multiple myeloma.

Mayo Clin Proc. 79: 867-874Huskey J.L. Heilman R.L. Khamash H. Fonseca R.

Kidney transplant in the Era of modern therapy for multiple myeloma.

Transplantation. 102: 1994-2001Stewart A.K. Rajkumar S.V. Dimopoulos M.A. et al.

Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma.

N Engl J Med. 372: 142-152Siegel D.S. Dimopoulos M.A. Ludwig H. et al.

Improvement in overall survival with carfilzomib, lenalidomide, and dexamethasone in patients with relapsed or refractory multiple myeloma.

J Clin Oncol. 36: 728-734Moreau P. Masszi T. Grzasko N. et al.

Oral ixazomib, lenalidomide, and dexamethasone for multiple myeloma.

N Engl J Med. 374: 1621-1634Carrier M. Le Gal G. Tay J. Wu C. Lee A.Y.

Rates of venous thromboembolism in multiple myeloma patients undergoing immunomodulatory therapy with thalidomide or lenalidomide: a systematic review and meta-analysis.

J Thromb Haemost. 9: 653-663Dimopoulos M.A. Jakubowiak A.J. McCarthy P.L. et al.

Developments in continuous therapy and maintenance treatment approaches for patients with newly diagnosed multiple myeloma.

Blood Cancer J. 10: 17Dimopoulos M.A. Gay F. Schjesvold F. et al.

Oral ixazomib maintenance following autologous stem cell transplantation (TOURMALINE-MM3): a double-blind, randomised, placebo-controlled phase 3 trial.

Lancet. 393: 253-264Jagannath S. Abonour R. Durie B.G.M. et al.

Impact of post-ASCT maintenance therapy on outcomes in patients with newly diagnosed multiple myeloma in Connect MM.

Blood Adv. 2: 1608-1615Lum E. Huang E. Bunnapradist S. Pham T. Danovitch G.

Acute kidney allograft rejection Precipitated by lenalidomide treatment for multiple myeloma.

Am J Kidney Dis. 69: 701-704Meyers D.E. Adu-Gyamfi B. Segura A.M. et al.

Fatal cardiac and renal allograft rejection with lenalidomide therapy for light-chain amyloidosis.

Am J Transpl. 13: 2730-2733Vaxman I. Eaton J. Lee H.E. Gertz M.A.

Acute liver rejection in a multiple myeloma patient treated with lenalidomide.

Case Rep Transpl. 2020: 8894922Walavalkar V. Adey D.B. Laszik Z.G. Jen K.-Y.

Severe renal allograft rejection resulting from lenalidomide therapy for multiple myeloma: case report.

Transplant Proc. 50: 873-876Everly M.J. Everly J.J. Susskind B. et al.

Bortezomib provides effective therapy for antibody- and cell-mediated acute rejection.

Transplantation. 86: 1754-1761Doberer K. Kläger J. Gualdoni G.A. et al.

CD38 antibody daratumumab for the treatment of Chronic active antibody-mediated kidney allograft rejection.

Transplantation. 105: 451-457Kwun J. Matignon M. Manook M. et al.

Daratumumab in Sensitized kidney transplantation: Potentials and Limitations of Experimental and clinical Use.

J Am Soc Nephrol. 30: 1206-1219

Anti-BCMA CAR T-cell therapy in multiple myeloma: can we do better?.

Leukemia. 34: 21-34Musto P. Anderson K.C. Attal M. et al.

Second primary malignancies in multiple myeloma: an overview and IMWG consensus.

Ann Oncol. 28: 228-245Yamasaki S. Yoshimoto G. Kohno K. et al.

Risk of secondary primary malignancies in multiple myeloma patients with or without autologous stem cell transplantation.

Int J Hematol. 109: 98-106Hickman L.A. Sawinski D. Guzzo T. Locke J.E.

Urologic malignancies in kidney transplantation.

Am J Transpl. 18: 13-22Casal M.A. Nolin T.D. Beumer J.H.

Estimation of kidney function in oncology: Implications for Anticancer drug selection and dosing.

CJASN. 14: 587-595Chancharoenthana W. Wattanatorn S. Vadcharavivad S. Eiam-Ong S. Leelahavanichkul A.

Agreement and Precision Analyses of various estimated Glomerular Filtration rate Formulae in cancer patients.

Sci Rep. 9: 19356

Proteinuria and hypertension with tyrosine kinase inhibitors.

Kidney Int. 80: 1271-1277Nadeau Nguyen M. Nayernama A. Jones S.C. Kasamon Y.L. Waldron P.E.

Solid organ transplant rejection associated with the Use of the immunomodulatory drugs (IMIDs).

Blood. 134: 2189Qualls D.A. Lewis G.D. Sanchorawala V. Staron A.

Orthotopic heart transplant rejection in association with immunomodulatory therapy for AL amyloidosis: a case series and review of the literature.

Am J Transpl. 19: 3185-3190Magnone M. Holley J.L. Shapiro R. et al.

Interferon-α-induced acute renal allograft rejection.

Transplantation. 59: 1068-1070Saab S. Kalmaz D. Gajjar N.A. et al.

Outcomes of acute rejection after interferon therapy in liver transplant recipients.

Liver Transplant. 10: 859-867LeBlanc R. Hideshima T. Catley L.P. et al.

Immunomodulatory drug costimulates T cells via the B7-CD28 pathway.

Blood. 103: 1787-1790Dutcher J.P. Schwartzentruber D.J. Kaufman H.L. et al.

High dose interleukin-2 (Aldesleukin) - expert consensus on best management practices-2014.

J ImmunoTherapy Cancer. 2: 26

The future of immune checkpoint therapy.

Science. 348: 56-61Lipson E.J. Bodell M.A. Kraus E.S. Sharfman W.H.

Successful administration of ipilimumab to two kidney transplantation patients with metastatic melanoma.

J Clin Oncol. 32: e69-e71Lipson E.J. Bagnasco S.M. Moore J. et al.

Tumor Regression and allograft rejection after administration of anti-PD-1.

N Engl J Med. 374: 896-898Ito T. Ueno T. Clarkson M.R. et al.

Analysis of the role of negative T cell Costimulatory pathways in CD4 and CD8 T cell-mediated Alloimmune responses in vivo.

J Immunol. 174: 6648-6656Yang J. Popoola J. Khandwala S. et al.

Critical role of donor tissue expression of programmed death ligand-1 in regulating cardiac allograft rejection and vasculopathy.

Circulation. 117: 660-669Riella L.V. Watanabe T. Sage P.T. et al.

Essential role of PDL1 expression on nonhematopoietic donor cells in acquired tolerance to vascularized cardiac allografts.

Am J Transpl. 11: 832-840Abdel-Wahab N. Safa H. Abudayyeh A. et al.

Checkpoint inhibitor therapy for cancer in solid organ transplantation recipients: an institutional experience and a systematic review of the literature.

J Immunother Cancer. 7: 106d’Izarny-Gargas T. Durrbach A. Zaidan M.

Efficacy and tolerance of immune checkpoint inhibitors in transplant patients with cancer: a systematic review.

Am J Transpl. 10: 2457-2465Murakami N. Mulvaney P. Danesh M. et al.

A multi-center study on safety and efficacy of immune checkpoint inhibitors in cancer patients with kidney transplant.

Kidney Int. 100: 196-205

National Cancer Institute (NCI). Immune Checkpoint Blockade for Kidney Transplant Recipients With Selected Unresectable or Metastatic Cancers. clinicaltrials.gov; 2021. Accessed September 25, 2021. https://clinicaltrials.gov/ct2/show/NCT03816332

Safety and efficacy of cemiplimab (PD-1 Blockade) in Selected Organ Transplant Recipients With Advanced Cutaneous Squamous Cell Carcinoma (CONTRAC).

clinicaltrials.gov, ()Barnett R. Barta V.S. Jhaveri K.D.

Preserved renal-allograft function and the PD-1 pathway inhibitor nivolumab.

N Engl J Med. 376: 191-192

ANZCTR - Registration. Accessed September 25, 2021. https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=372928

Adam B. Murakami N. Reid G. et al.

Gene expression Profiling in kidney transplants with immune checkpoint inhibitor-associated adverse Events.

Clin J Am Soc Nephrol. 16: 1376-1386Hurkmans D.P. Verhoeven J.G.H.P. de Leur K. et al.

Donor-derived cell-free DNA detects kidney transplant rejection during nivolumab treatment.

J Immunother Cancer. 7: 182Lakhani L. Alasfar S. Bhalla A. et al.

Utility of serial donor-derived cell-free DNA measurements for detecting allograft rejection in a kidney transplant recipient after PD-1 checkpoint inhibitor administration.

Transpl Direct. 7: e656Duni A. Kitsos A. Liapis G. Tatsis V. Pappas C. Dounousi E.

Acute kidney transplant rejection after administration of nivolumab in a dialysis patient with a failed graft.

Kidney Int Rep. 6: 1459-1463Mejia C.D. Frank A.M. Singh P. Yadav A.

Immune checkpoint inhibitor therapy-associated graft intolerance syndrome in a failed kidney transplant recipient.

Am J Transpl. 21: 1322-1325Kitchlu A. Jhaveri K.D. Sprangers B. Yanagita M. Wanchoo R.

Immune-checkpoint inhibitor use in patients with end-stage kidney disease: an analysis of reported cases and literature review.

Clin Kidney J. 14 (): 2012-2022Dinh A.R. Wong S.W. Martin T.G. Wolf J.L. Webber A.B.

Outcomes of kidney transplant recipients with ESKD due to plasma cell dyscrasia: a case series.

Clin Transplant. 36e14541Schreiber B. Abdelrahim M. Abudayyeh A. Murakami N.

Emerging concepts in managing malignancy in kidney transplant patients..

Semin Nephrol. 42: 63-75

留言 (0)

沒有登入
gif