Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts

Jessica Hartmann, Martina Schüßler-Lenz, Attilio Bondanza, Christian J Buchholz, Jessica Hartmann, Martina Schüßler-Lenz, Attilio Bondanza, Christian J Buchholz

Abstract

Chimeric antigen receptor (CAR) T cell therapy, together with checkpoint inhibition, has been celebrated as a breakthrough technology due to the substantial benefit observed in clinical trials with patients suffering from relapsed or refractory B-cell malignancies. In this review, we provide a comprehensive overview of the clinical trials performed so far worldwide and analyze parameters such as targeted antigen and indication, CAR molecular design, CAR T cell manufacturing, anti-tumor activities, and related toxicities. More than 200 CAR T cell clinical trials have been initiated so far, most of which aim to treat lymphoma or leukemia patients using CD19-specific CARs. An increasing number of studies address solid tumors as well. Notably, not all clinical trials conducted so far have shown promising results. Indeed, in a few patients CAR T cell therapy resulted in severe adverse events with fatal outcome. Of note, less than 10% of the ongoing CAR T cell clinical trials are performed in Europe. Taking lead from our analysis, we discuss the problems and general hurdles preventing efficient clinical development of CAR T cells as well as opportunities, with a special focus on the European stage.

Keywords: ATMPs; cancer; immunotherapy; regulatory issues; toxicities.

© 2017 The Authors. Published under the terms of the CC BY 4.0 license.

Figures

Figure 1. CAR T cell therapy—principle and…
Figure 1. CAR T cell therapy—principle and clinical trial overview
(A) The CAR T cell therapy process. T cells are isolated from blood of the patient or a donor, activated, and then genetically engineered to express the CAR construct (an example shown in gray above the vector particle in violet). After ex vivo expansion of the CAR T cells, they are formulated into the final product. The patient undergoes either a conditional chemotherapy or the CAR T cell product is directly infused. (B) Schematic representation of a T cell receptor (TCR) and four types of chimeric antigen receptors (CARs) being displayed on the surface of a T cell while contacting their antigen (red) on the tumor cell. The single‐chain variable fragment (scFv) as ligand‐binding domain mediating tumor cell recognition in CARs is shown in light blue with the VH and VL domains being connected via a long flexible linker and transmembrane domain to intracellular signaling domains. Pro‐inflammatory cytokines or co‐stimulatory ligands expressed by the CAR T cells are depicted for the 4th generation. (C) Overview of so‐called smart CAR T cells products. Pooled CAR T cell products consist of two or more single‐targeting CAR T cell types with distinct antigen specificities. Multi‐CAR T cells harbor several CAR molecules with different antigen specificities. A tandem CAR T cell expresses a CAR construct harboring two ligand‐binding domains with different antigen specificities. In a conditional CAR T cell activation and co‐stimulation are separated on two CAR constructs recognizing different target antigens. In the split CAR construct the ligand‐binding or signaling domain is physically separated allowing controlled CAR T cell activation. iCAR T cells additionally express a receptor engineered to recognize an antigen expressed on normal tissue to provide an inhibitory signal in turn. In addition CAR T cells can be equipped with suicide genes or switches (e.g., iCasp9) allowing ablation of CAR T cells. (D) Left, status of published CAR T cell gene therapy trials or trials registered at ClinicalTrials.gov including long‐term follow‐up studies. The status of one trial is unknown and not listed. The total number of clinical trials (dark blue bars) is compared to published clinical trials (light blue bars). The asterisk indicates zero trials. Right, phases of CAR T cell gene therapy trials. Long‐term follow‐up studies are not included. For nine trials, the phase classification is unknown. The asterisk indicates zero trials.
Figure 2. CAR T cell trials over…
Figure 2. CAR T cell trials over time and geographical distribution
(A) Timeline of cancer CAR T cell trials as listed in Datasets EV1 and EV2 distinguishing between ongoing number (dark blue bars) and newly initiated trials in the indicated year (light blue bars). (B) Geographical distribution of worldwide ongoing CAR T cells clinical trials (left) and distribution of trial sites of the ongoing European studies (right). Five studies are multi‐centric, of which four are multi‐country trials in Europe (Dataset EV5). Long‐term follow‐up studies are not included. Color code indicates the prevalence of trials from low (green) to high (red).
Figure 3. Indication, age, CAR generation, and…
Figure 3. Indication, age, CAR generation, and targeted antigen in clinical trials
(A) Solid tumors versus tumors of the hematopoietic and lymphoid system. The number of ongoing trials (dark blue bar) is compared to the number of non‐active trials (light blue bar). (B) Patient age distribution for solid tumors (light blue bars) and hematological malignancies (dark blue bars). (C) Generation of the CAR constructs applied. (D) Targeted antigens separated for tumors of hematopoietic or lymphoid origin (upper panel) and for solid tumors (lower panel).
Figure 4. CAR T cell dose and…
Figure 4. CAR T cell dose and percentages of CAR‐positive cells within CAR T cell products
(A, B) CAR T cell dose indicated in the study description of published CAR T cell gene therapy trials or trials registered at ClinicalTrials.gov. The CAR T cell dose is normalized to 75 kg or 1.72 m2 per dose. CAR T cells are either administered as a fixed dose (A) or in a dose escalation regimen (B). Each dot represents a single trial. (C, D) The reported number of CAR‐positive cells as given in Datasets EV3 and EV4 in column “%CAR+ cells (median; range)” was used to identify the median amount of CAR‐positive T cells in the various cell products within one clinical trial (C) and the range of variability between cell products within one clinical trial (lowest (min) and highest (max) percentage CAR‐positive T cells per trial) (D).
Figure 5. Clinical outcome
Figure 5. Clinical outcome
(A, B) Best clinical outcome for hematological malignancies (A) and solid tumors (B) dependent on the targeted antigen. The number of treated patients is provided in brackets below the targeted antigen. CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; NR, no response; NE, not evaluable.
Figure 6. Important drivers in CAR T…
Figure 6. Important drivers in CAR T cell trials

References

    1. Abou‐El‐Enein M, Schneider CK (2016) Deciphering the EU clinical trials regulation. Nat Biotechnol 34: 231–233
    1. Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, Liu E, Dakhova O, Ashoori A, Corder A et al (2015) Human epidermal growth factor receptor 2 (HER2) ‐specific chimeric antigen receptor‐modified T cells for the immunotherapy of HER2‐positive sarcoma. J Clin Oncol 33: 1688–1696
    1. Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G, Chew A, Zhao Y, Levine BL, Albelda SM et al (2014) Mesothelin‐specific chimeric antigen receptor mRNA‐engineered T cells induce anti‐tumor activity in solid malignancies. Cancer Immunol Res 2: 112–120
    1. Brentjens RJ, Riviere I, Park JH, Davila ML, Wang X, Stefanski J, Taylor C, Yeh R, Bartido S, Borquez‐Ojeda O et al (2011) Safety and persistence of adoptively transferred autologous CD19‐targeted T cells in patients with relapsed or chemotherapy refractory B‐cell leukemias. Blood 118: 4817–4828
    1. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C, Olszewska M et al (2013) CD19‐targeted T cells rapidly induce molecular remissions in adults with chemotherapy‐refractory acute lymphoblastic leukemia. Sci Transl Med 5: 177ra38
    1. Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang W‐C, Naranjo A, Starr R, Wagner J, Wright C et al (2015) Bioactivity and safety of IL13Ralpha2‐redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glioblastoma. Clin Cancer Res 21: 4062–4072
    1. Casucci M, Di Nicolis Robilant B, Falcone L, Camisa B, Norelli M, Genovese P, Gentner B, Gullotta F, Ponzoni M, Bernardi M et al (2013) CD44v6‐targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood 122: 3461–3472
    1. Chmielewski M, Abken H (2015) TRUCKs: the fourth generation of CARs. Expert Opin Biol Ther 15: 1145–1154
    1. Cicalese MP, Aiuti A (2015) Clinical applications of gene therapy for primary immunodeficiencies. Hum Gene Ther 26: 210–219
    1. Cruz CRY, Micklethwaite KP, Savoldo B, Ramos CA, Lam S, Ku S, Diouf O, Liu E, Barrett AJ, Ito S et al (2013) Infusion of donor‐derived CD19‐redirected virus‐specific T cells for B‐cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood 122: 2965–2973
    1. Dai H, Zhang W, Li X, Han Q, Guo Y, Zhang Y, Wang Y, Wang C, Shi F, Zhang Y et al (2015) Tolerance and efficacy of autologous or donor‐derived T cells expressing CD19 chimeric antigen receptors in adult B‐ALL with extramedullary leukemia. Oncoimmunology 4: e1027469
    1. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez‐Ojeda O, Olszewska M et al (2014) Efficacy and toxicity management of 19‐28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 6: 224ra25
    1. DeFrancesco L (2016) Juno's wild ride. Nat Biotechnol 34: 793
    1. DeFrancesco L (2017) CAR‐T's forge ahead, despite Juno deaths. Nat Biotechnol 35: 6–7
    1. Duda GN, Grainger DW, Frisk ML, Bruckner‐Tuderman L, Carr A, Dirnagl U, Einhaupl KM, Gottschalk S, Gruskin E, Huber C et al (2014) Changing the mindset in life sciences toward translation: a consensus. Sci Transl Med 6: 264cm12
    1. Fesnak AD, June CH, Levine BL (2016) Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer 16: 566–581
    1. Fitzgerald JC, Weiss SL, Maude SL, Barrett DM, Lacey SF, Melenhorst JJ, Shaw P, Berg RA, June CH, Porter DL et al (2017) Cytokine Release Syndrome After Chimeric Antigen Receptor T Cell Therapy for Acute Lymphoblastic Leukemia. Crit Care Med 45: e124–e131
    1. Grupp SA, Laetsch TW, Buechner J, Bittencourt H, Maude SL, Verneris MR, Myers GD, Boyer MW, Rives S, Moerloose BD et al (2016) Analysis of a global registration trial of the efficacy and safety of CTL019 in pediatric and young adults with relapsed/refractory acute lymphoblastic leukemia (ALL). Blood 128: 221
    1. Guo B, Chen M, Han Q, Hui F, Dai H, Zhang W, Zhang Y, Wang Y, Zhu H, Han W (2016) CD138‐directed adoptive immunotherapy of chimeric antigen receptor (CAR)‐modified T cells for multiple myeloma. J Cell Immunother 2: 28–35
    1. Jensen MC, Popplewell L, Cooper LJ, DiGiusto D, Kalos M, Ostberg JR, Forman SJ (2010) Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19‐specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant 16: 1245–1256
    1. Kantarjian H, Stein A, Gokbuget N, Fielding AK, Schuh AC, Ribera J‐M, Wei A, Dombret H, Foa R, Bassan R et al (2017) Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med 376: 836–847
    1. Katz SC, Burga RA, McCormack E, Wang LJ, Mooring W, Point GR, Khare PD, Thorn M, Ma Q, Stainken BF et al (2015) Phase I hepatic immunotherapy for metastases study of intra‐arterial chimeric antigen receptor‐modified T‐cell therapy for CEA+ liver metastases. Clin Cancer Res 21: 3149–3159
    1. Kebriaei P, Singh H, Huls MH, Figliola MJ, Bassett R, Olivares S, Jena B, Dawson MJ, Kumaresan PR, Su S et al (2016) Phase I trials using sleeping beauty to generate CD19‐specific CAR T cells. J Clin Investig 126: 3363–3376
    1. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich JR, Canevari S, Rogers‐Freezer L et al (2006) A phase I study on adoptive immunotherapy using gene‐modified T cells for ovarian cancer. Clin Cancer Res 12: 6106–6115
    1. Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, Hakim FT, Halverson DC, Fowler DH, Hardy NM et al (2013) Donor‐derived CD19‐targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 122: 4129–4139
    1. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RPT, Carpenter RO, Stetler‐Stevenson M, Yang JC, Phan GQ, Hughes MS, Sherry RM et al (2015) Chemotherapy‐refractory diffuse large B‐cell lymphoma and indolent B‐cell malignancies can be effectively treated with autologous T cells expressing an anti‐CD19 chimeric antigen receptor. J Clin Oncol 33: 540–549
    1. Kohn DB (2010) Update on gene therapy for immunodeficiencies. Clin Immunol 135: 247–254
    1. Koneru M, O'Cearbhaill R, Pendharkar S, Spriggs DR, Brentjens RJ (2015) A phase I clinical trial of adoptive T cell therapy using IL‐12 secreting MUC‐16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. J Transl Med 13: 102
    1. Lamers CHJ, Sleijfer S, Vulto AG, Kruit WHJ, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E (2006) Treatment of metastatic renal cell carcinoma with autologous T‐lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol 24: e20–e22
    1. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, Grupp SA, Mackall CL (2014) Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124: 188–195
    1. Lee DW, Kochenderfer JN, Stetler‐Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN et al (2015) T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose‐escalation trial. Lancet 385: 517–528
    1. Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hosing CM, Ghobadi A, Budde LE, Bot A, Rossi JM et al (2017) Phase 1 results of ZUMA‐1: a multicenter study of KTE‐C19 Anti‐CD19 CAR T cell therapy in refractory aggressive lymphoma. Mol Ther 25: 285–295
    1. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, Rossig C, Russell HV, Diouf O, Liu E et al (2011) Antitumor activity and long‐term fate of chimeric antigen receptor‐positive T cells in patients with neuroblastoma. Blood 118: 6050–6056
    1. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF et al (2014) Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 371: 1507–1517
    1. Mock U, Nickolay L, Philip B, Cheung GW‐K, Zhan H, Johnston ICD, Kaiser AD, Peggs K, Pule M, Thrasher AJ et al (2016) Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS prodigy. Cytotherapy 18: 1002–1011
    1. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA (2010) Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther 18: 843–851
    1. Mullard A (2017) PRIME time at the EMA. Nat Rev Drug Discov 16: 226–228
    1. Neelapu SS, Locke FL, Bartlett NL, Lekakis L, Miklos D, Lin Y, Timmerman J, Stiff PJ, Friedberg J, Flinn I et al (2016) KTE‐C19 (anti‐CD19 CAR T Cells) Induces Complete Remissions in Patients with Refractory Diffuse Large B‐Cell Lymphoma (DLBCL): Results from the Pivotal Phase 2 ZUMA‐1. ASH 58th Annual Meeting & Exposition
    1. Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, Meechoovet HB, Bautista C, Chang W‐C, Ostberg JR et al (2007) Adoptive transfer of chimeric antigen receptor re‐directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther 15: 825–833
    1. Park JH, Rivere I, Wang X, Senechal B, Curran K, Sauter C, Wang Y, Santomasso B, Li D, Brentjens R et al (2017) Impact of disease burden and transplant on long‐term survival after CD19 CAR therapy in adults with relapsed B‐cell acute lymphoblastic leukemia: Abstract CT078. AACR Annual Meeting 2017
    1. Passweg JR, Baldomero H, Bader P, Bonini C, Cesaro S, Dreger P, Duarte RF, Dufour C, Kuball J, Farge‐Bancel D et al (2016) Hematopoietic stem cell transplantation in Europe 2014: more than 40,000 transplants annually. Bone Marrow Transplant 51: 786–792
    1. Petrausch U, Schuberth PC, Hagedorn C, Soltermann A, Tomaszek S, Stahel R, Weder W, Renner C (2012) Re‐directed T cells for the treatment of fibroblast activation protein (FAP)‐positive malignant pleural mesothelioma (FAPME‐1). BMC Cancer 12: 615
    1. Poirot L, Philip B, Schiffer‐Mannioui C, Le Clerre D, Chion‐Sotinel I, Derniame S, Potrel P, Bas C, Lemaire L, Galetto R et al (2015) Multiplex genome‐edited T‐cell manufacturing platform for “Off‐the‐Shelf” adoptive T‐cell immunotherapies. Can Res 75: 3853–3864
    1. Porter DL, Hwang W‐T, Frey NV, Lacey SF, Shaw PA, Loren AW, Bagg A, Marcucci KT, Shen A, Gonzalez V et al (2015) Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med 7: 303ra139
    1. Priesner C, Aleksandrova K, Esser R, Mockel‐Tenbrinck N, Leise J, Drechsel K, Marburger M, Quaiser A, Goudeva L, Arseniev L et al (2016) Automated enrichment, transduction and expansion of clinical‐scale CD62L+ T cells for manufacturing of GTMPs. Hum Gene Ther 27: 860–869
    1. Qasim W, Zhan H, Samarasinghe S, Adams S, Amrolia P, Stafford S, Butler K, Rivat C, Wright G, Somana K et al (2017) Molecular remission of infant B‐ALL after infusion of universal TALEN gene‐edited CAR T cells. Sci Transl Med 9: eaaj2013
    1. Ramos CA, Savoldo B, Torrano V, Ballard B, Zhang H, Dakhova O, Liu E, Carrum G, Kamble RT, Gee AP et al (2016) Clinical responses with T lymphocytes targeting malignancy‐associated kappa light chains. J Clin Investig 126: 2588–2596
    1. Renner M, Anliker B, Sanzenbacher R, Schuele S (2015) Regulation of clinical trials with advanced therapy medicinal products in germany. Adv Exp Med Biol 871: 87–101
    1. Rietschel ET, Bruckner‐Tuderman L, Schutte G, Wess G (2015) Moving medicine forward faster. Sci Transl Med 7: 277ed2
    1. Sant M, Allemani C, Tereanu C, de Angelis R, Capocaccia R, Visser O, Marcos‐Gragera R, Maynadie M, Simonetti A, Lutz J‐M et al (2010) Incidence of hematologic malignancies in Europe by morphologic subtype: results of the HAEMACARE project. Blood 116: 3724–3734
    1. Schimmer J, Breazzano S (2016) Investor outlook: rising from the ashes: GSK's European approval of strimvelis for ADA‐SCID. Hum Gene Ther Clin Dev 27: 57–61
    1. Till BG, Jensen MC, Wang J, Chen EY, Wood BL, Greisman HA, Qian X, James SE, Raubitschek A, Forman SJ et al (2008) Adoptive immunotherapy for indolent non‐Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20‐specific T cells. Blood 112: 2261–2271
    1. Till BG, Jensen MC, Wang J, Qian X, Gopal AK, Maloney DG, Lindgren CG, Lin Y, Pagel JM, Budde LE et al (2012) CD20‐specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4‐1BB domains: pilot clinical trial results. Blood 119: 3940–3950
    1. Turtle CJ, Hanafi L‐A, Berger C, Gooley TA, Cherian S, Hudecek M, Sommermeyer D, Melville K, Pender B, Budiarto TM et al (2016) CD19 CAR‐T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Investig 126: 2123–2138
    1. Walker A, Johnson R (2016) Commercialization of cellular immunotherapies for cancer. Biochem Soc Trans 44: 329–332
    1. Wang Y, Zhang W‐Y, Han Q‐W, Liu Y, Dai H‐R, Guo Y‐L, Bo J, Fan H, Zhang Y, Zhang Y‐J et al (2014) Effective response and delayed toxicities of refractory advanced diffuse large B‐cell lymphoma treated by CD20‐directed chimeric antigen receptor‐modified T cells. Clin Immunol 155: 160–175
    1. Wang X, Popplewell LL, Wagner JR, Naranjo A, Blanchard MS, Mott MR, Norris AP, Wong CW, Urak RZ, Chang W‐C et al (2016) Phase 1 studies of central memory‐derived CD19 CAR T‐cell therapy following autologous HSCT in patients with B‐cell NHL. Blood 127: 2980–2990
    1. Whitmore KV, Gaspar HB (2016) Adenosine deaminase deficiency – more than just an immunodeficiency. Front Immunol 7: 314
    1. Xu Y, Zhang M, Ramos CA, Durett A, Liu E, Dakhova O, Liu H, Creighton CJ, Gee AP, Heslop HE et al (2014) Closely related T‐memory stem cells correlate with in vivo expansion of CAR.CD19‐T cells and are preserved by IL‐7 and IL‐15. Blood 123: 3750–3759
    1. You F, Jiang L, Zhang B, Lu Q, Zhou Q, Liao X, Wu H, Du K, Zhu Y, Meng H et al (2016) Phase 1 clinical trial demonstrated that MUC1 positive metastatic seminal vesicle cancer can be effectively eradicated by modified Anti‐MUC1 chimeric antigen receptor transduced T cells. Sci China Life Sci 59: 386–397
    1. Zhang T, Cao L, Xie J, Shi N, Zhang Z, Luo Z, Yue D, Zhang Z, Wang L, Han W et al (2015) Efficiency of CD19 chimeric antigen receptor‐modified T cells for treatment of B cell malignancies in phase I clinical trials: a meta‐analysis. Oncotarget 6: 33961–33971
    1. Zhang Y, Zhang W, Dai H, Wang Y, Shi F, Wang C, Guo Y, Liu Y, Chen M, Feng K et al (2016) An analytical biomarker for treatment of patients with recurrent B‐ALL after remission induced by infusion of anti‐CD19 chimeric antigen receptor T (CAR‐T) cells. Sci China Life Sci 59: 379–385
    1. Zhang E, Xu H (2017) A new insight in chimeric antigen receptor‐engineered T cells for cancer immunotherapy. J Hematol Oncol 10: 1
    1. Zhao Z, Condomines M, van der Stegen SJC, Perna F, Kloss CC, Gunset G, Plotkin J, Sadelain M (2015) Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T Cells. Cancer Cell 28: 415–428

Source: PubMed

3
Se inscrever