Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Transfusion Medicine Reviews 33 (2019) 98–110

Contents lists available at ScienceDirect

Transfusion Medicine Reviews


journal homepage:
https://www.journals.elsevier.com/transfusion-medicine-reviews/

Risks and Benefits of Chimeric Antigen Receptor T-Cell (CAR-T) Therapy


in Cancer: A Systematic Review and Meta-Analysis☆
Emma J.M. Grigor a,b,c,1, Dean Fergusson b,c,f,1, Natasha Kekre d, Joshua Montroy b,c, Harold Atkins d,e,
Matthew D. Seftel j, Mads Daugaard k,l, Justin Presseau b,c, Kednapa Thavorn b,c,o, Brian Hutton c,g,
Robert A. Holt i,m,n, Manoj M. Lalu a,b,h,⁎,1
a
Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
b
Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
c
School of Epidemiology and Pubic Health, University of Ottawa, Ottawa, Ontario, Canada
d
Blood and Marrow Transplant Program, The Ottawa Hospital, Ottawa, Ontario, Canada
e
Cancer Therapeutic Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
f
Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
g
Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
h
Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
i
Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
j
Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba
k
Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
l
Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
m
Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
n
Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
o
Institute for Clinical Evaluative Sciences, Ottawa, Ontario, Canada

a r t i c l e i n f o a b s t r a c t

Available online 14 February 2019 Promising efficacy results of chimeric antigen receptor (CAR) T-cell therapy have been tempered by safety con-
siderations. Our objective was to comprehensively summarize the efficacy and safety of CAR-T cell therapy in pa-
Keywords: tients with relapsed or refractory hematologic or solid malignancies. MEDLINE, Embase, and the Cochrane
CAR-T cell therapy Register of Controlled Trials (inception – November 21, 2017). Interventional studies investigating CAR-T cell
Systematic review
therapy in patients with malignancies were included. Our primary outcome of interest was complete response
Cancer
(defined as the absence of detectable cancer). Two independent reviewers extracted relevant data, assessed
Hematologic
risk of bias, and graded the quality of evidence using established methods. A total of 42 hematological malignancy
studies and 18 solid tumor studies met were included (913 participants). Of 486 evaluable hematologic patients,
54.4% [95% CI, 42.5%-65.9%] experienced complete response in 27 CD19 CAR-T cell therapy studies. Of 65
evaluable hematologic patients, 24.4% [95% CI, 9.4%-50.3%] experienced complete response in seven non-CD19
CAR-T cell therapy studies. Cytokine release syndrome was experienced by 55.3% [95% CI, 40.3%-69.4%] of pa-
tients and neurotoxicity 37.2% [95% CI, 28.6%-46.8%] of patients with hematologic malignancies. Of 86 evaluable
solid tumor patients, 4.1% [95% CI, 1.6%-10.6%] experienced complete response in eight CAR-T cell therapy stud-
ies. Limitations include heterogeneity of study populations, as well as high risk of bias of included studies. There
was a strong signal for efficacy of CAR-T cell therapy in patients with CD19+ hematologic malignancies and no
overall signal in solid tumor trials published to date. These results will help inform patients, physicians, and
other stakeholders of the benefits and risks associated with CAR-T cell therapy.
© 2019 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).

Abbreviations: ALL, acute lymphocytic leukemia; AML, acute myeloid leukemia; BioCanRx, Biotherapeutics for Cancer Treatment; CAR-T, chimeric antigen receptor T cell; CD, cluster of
differentiation; CLL, chronic lymphocytic leukemia; DistillerSR, Distiller Systematic Review; GRADE, grading of recommendations, assessment, development and evaluations; IHE, Institute
of Health Economics; NHL, non-Hodgkin's lymphoma; RECIST, response evaluation criteria in solid tumors.
☆ Systematic review registration: PROSPERO CRD42017075331.
⁎ Corresponding author at: Manoj M Lalu, MD, PhD, 501 Smyth Road, PO Box 201B, Ottawa, ON, K1H 8L6, Canada.
E-mail addresses: manojlalu@gmail.com, mlalu@toh.ca (M.M. Lalu).
@manojlalu (M.M. Lalu).
1
Authors contributed equally to work.

https://doi.org/10.1016/j.tmrv.2019.01.005
0887-7963/© 2019 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110 99

Contents

Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99
Data sources and searches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
Study selection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
Data extraction and quality assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
Data synthesis and analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
Role of the funding source . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
Results . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
Declaration of Interests . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
Appendix A. Supplementary data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108

Chimeric antigen receptor (CAR) T cell therapy is a cell-based gene We present a comprehensive, up-to-date and methodologically rig-
therapy that has been used to treat patients with relapsed or refractory orous systematic review and meta-analysis summarizing the benefits
malignancies in several phase I and phase II clinical trials. This individu- and risks of CAR-T cell therapy in patients with relapsed or refractory
alized patient therapy harnesses the natural function of the body's T hematological malignancies and solid tumors from all published clinical
lymphocytes. T cells from either a donor or the recipient are genetically trials. This summary will help inform decisions by patients, clinicians
engineered to express a specific CAR (eg, using a retrovirus) designed to and other stakeholders.
recognize a specific tumor associated antigen; this allows the T-cells to ac-
tively target and selectively kill cells expressing that antigen (eg, CD19,
HER-2). Early clinical trials that investigated anti-CD19 CAR-T cells in he- Methods
matologic malignancies have demonstrated continued remission in pa-
tients with late stage disease [1-5]. CAR-T cell therapy investigating This systematic review and meta-analysis follows the Preferred
different cancer-specific targets have also shown some potential in stud- Reporting Items for Systematic Reviews and Meta-analyses guidelines,
ies of non-hematopoietic malignancies [6]. These promising results have as shown in the appendix (Appendix 1) [9]. The protocol is available
been tempered by safety considerations, including cytokine release syn- in the International Prospective Register of Systematic Reviews
drome, neurotoxicity, and other rare and uncommon adverse events [7,8]. (CRD42017075331). The protocol has been previously published [10].

1637 Solid cancer records identified through 1674 Blood cancer records identified through
database searching database searching

1356 = Solid 1381 = Blood


1518 Solid cancer records screened after 1492 Blood cancer records screened after
cancer cancer
duplicates removed duplicates removed
excluded excluded
reports at reports at
title/abstract title/abstract

144 Solid cancer excluded 162 Solid cancer full-text reports assessed 69 Blood cancer excluded reports
reports
111 Blood cancer full-text reports assessed 39 commentary, editorials,
96 pre-clinical or animal or letters, or reviews
manufacturer studies 5 Duplicate
41 commentary, editorials, 2 Partial duplicate
letters, or reviews 4 pre-clinical or animal or
3 Duplicates manufacturer studies
3 other 4 abstract
1 Abstract 3 did not report outcomes of
interest
12 other (including protocols)

18 Solid cancer studies were included

42 Blood cancer studies were included

Fig 1. PRISMA diagram summarizing screening and selection of studies included in review.
100 E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110

Table 1
Characteristics of the included studies and participants

First author (year) Country Trial Phase Patients evaluated Age, years mean (range) Cancer type(s) Follow-up
n (% male)

Hematologic malignancies
Till 2008 [25] United States I 9 (89) 60.2 (43–77) NHL NP
Kalos 2011 [26] United States I 3 (100) 68.7 (64–77) ALL 15 m
Brentjens 2011 [27] United States I 8 (80) 63.9 (51–73) ALL, CLL NP
Till 2012 [28] United States I 4 (100) 59.2 (28–80) NHL NP
Kochenderfer 2012 [29] United States I/II 8 (NP) 55.8 (47–63) NHL, CLL NP
Brentjens 2013 [30] United States I 5 (80) 52.4 (23–66) ALL NP
Cruz 2013 [31] United States I 8 (63) 38.5 (9–59) ALL, CLL NP
Kochenderfer 2013 [32] United States I 10 (80) 52.4 (44–66) NHL, CLL NP
Ritchie 2013 [33] Australia I 5 (33) 70.6 (64–78) AML NP
Davila 2014 [4] United States I 16 (75) 50 (NP)b ALL NP
Maude 2014 [23] United States I/IIa 30 (60) 8.7 (8–12) ALL NP
Wang 2014 [34] China I/II 7 (85) 62.4 (37–85) NHL 14 m
Dai 2015 [35] China NP 9 (44) 38.9 (15–65) ALL NP
Lee 2015 [3] United States I 21 (66) 14.7 (5–27) NHL, ALL NP
Porter 2015 [36] United States I 14 (85) 66 (51–78)b CLL 19 m (6–53)
Wang 2015 [37] China I/II 1 (100) 41 (NP) AML NP
Brudno 2016 [38] United States I 20 (55) 48 (20–68) NHL, ALL, CLL 6m
Cai 2016 [39] China I 1 (0) 71 (NP) ALL 31 d
Hu 2016 [40] China NP 1 (0) 43 (NP) ALL 32 w
Kebriaei 2016 [41] United States I 26 (NP) 40 (21–61) NHL, ALL NP
Locke 2016 [42] United States I 7 (NP) NP NHL NP
Prudent 2016 [43] United States NP 2 (50) 29 (25–33) ALL NP
Turtle 2016a [44] United States I/II 30 (NP) 40 (20–73)b ALL NP
Turtle 2016b [45] United States I 32 (84) 57 (22–70) NHL NP
Wang 2016 [46] United States I 16 (56) 60 (23–75) NHL 25.8 m (24–37)
Zhu 2016 [47] China NP 2 (50) 34 (29–39) ALL NP
Chen 2017 [48] China I/II 6 (17) 26.5 (8–44) ALL 14 m
Fitzgerald 2017 [49] United States I/IIa 39 (51) 11 (5–22)b ALL 28 d
Fry 2017 [50] United States I 21 (62) 19 (7–30)b ALL 21 m
Gardner 2017 [51] United States I 45 (51) 12.2 (1.3–25.3)b ALL 9.6 m (2–28)
Hay 2017 [52] United States I/II 133 (70) 54 (20–73)b CLL, ALL, NHL 30 d
Kochenderfer 2017 [53] United States I/II 22 (NP) 47.4 (38–64) NHL 26 m
Lai 2017 [54] China NP 1 (NP) NP ALL 80 d
Neelapu 2017 [55] United States II 28 (64) 58 (23–76) NHL 15.4 m
Pan 2017 [56] China NP 51 (63) 2–68 ALL 206 d (45–427)
Qasim 2017 [57] United Kingdom 0 2 (0) 1.1 (0.9–1.3) ALL 15 m (12–18)
Ramos 2017 [58] United States I 9 (67) 34.6 (20–65) Hodgkin lymphoma 15 m
Rossig 2017 [59] Germany I/II 11 (83) 9 (2–12) ALL 12 m (1–37)
Schuster 2017 [60] United States II 92 (61) 25–77 NHL 28.6 m (3.5–3.79)
Smith 2017 [61] United States NP 1 (100) 21 (NP) ALL 28 d
Wang 2017 [62] China I 18 (72) 33 (13–77)b NHL ≥2m
Zahid 2017 [63] United States NP 1 (100) 65 (NP) NHL 17 m

Solid malignancies
Morgan 2010 [64] United States I/II 1 (0) 39 Colon cancer NP
Maus 2013 [8] United States I 3 (NP) NP Pancreatic exceadenocarcinoma NP
Ahmed 2015 [65] United States I/II 19 (47) 17 (7.7–29.6)b Sarcoma, neuroectodermal tumor, DSRCT 10.2 m (1.1–37)
Brown 2015 [66] United States I 3 (NP) NP Glioblastoma NP
Katz 2015 [67] United States I 6 (67) 57 (51–66) Liver metastases NP
Brown 2016 [68] United States I 1 (100) 50 Glioblastoma 298 d
Feng 2016 [69] China I 11 (45) 58 (40–66)b NSCLC NP
Junghans 2016 [70] United States I 5 (100) 61 (51–75)b Prostate cancer NP
Lamers 2016 [71] The Netherlands I/II 12 (NP) NP MRCC 9.5 m
You 2016 [72] United States I 1 (100) NP Metastatic seminal vesicle cancer NP
Feng 2017a [73] China I 1 (0) 52 Advanced cholangio-carcinoma NP
Feng 2017b [74] China I 11 (82) 60.5 (50–75) Biliary tract and pancreatic cancers 4.8 m (1.5–8.3)
Hege 2017 [75] United States I 23 (NP) NP colorectal cancer 20 d
O'Rourke 2017 [76] United States I 10 (50) 59.5 (45–76)b Glioblastoma NP
Tanyi 2017 [77] United States NP 1 (0) 52 (NP) ovarian cancer 29 d
Tchou 2017 [78] United Kingdom 0 6 (0) 55.3 (44–64) metastatic breast cancer 10 m (3–28)
Thistlethwaite 2017 [79] United Kingdom I 14 (57) 47.7 (36–66) CEA+ tumors of GI tract 56 m
Zhang 2017 [80] United States I 10 (70) 58 (48.8–67)b metastatic colorectal cancer 77 m

Abbreviations: ALL, acute lymphocytic leukemia; AML, acute myeloid leukemia; CEA, carcinoembryonic antigen; CLL, chronic lymphocytic leukemia; d, days; DSRCT, desmoplastic small
round cell tumor; GI, gastrointestinal; mo, months; n, number; NHL, non-Hodgkin's lymphoma; NP, not provided; MRCC, metastatic renal cell carcinoma; NSCLC, non-small cell lung can-
cer; w, weeks; y, year.

Data sources and searches and abstracts were excluded. MEDLINE (OVID interface, including In-
Process and Epub Ahead of Print) Embase (OVID interface), and the
Uncontrolled or controlled interventional studies that investigated Cochrane Central Register of Controlled Trials (Wiley interface) data-
CAR-T cell therapy in patients with refractory or relapsed hematologic bases were search from inception to November 21, 2017. Two separate
or solid malignancies were considered. Only studies that reported out- search strategies were developed for hematological malignancies and
comes of interest were included. Gray literature, commentaries, letters, solid tumors using key words related to CAR-T cell therapy (search
E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110 101

Table 2
Chimeric Antigen Receptor T-cell Therapy Characteristics of the Included Studies

First author T-cell Transfection/ CAR-T dose Previous treatments Lymphodepletion Co-stim CAR antigen
(year) origin transduction method (cells/m2 or cells/kg) domain target

Hematologic malignancies
Till 2008 [25] Auto Electroporation 1×108–3.3×108/m2 Chemo Chemo NC CD20
Kalos 2011 [26] Auto Retrovirus 1.46×105–1.6×107/kg Biologic, chemo Chemo (Cy, Fl, Pen, Be, Le), 4-1BB CD19
biologic (All, Ri)
Brentjens 2011 Auto NP 1.2×107–3×107/kg Chemo Chemo (Cy) CD28 CD19
[27]
6 7
Till 2012 [28] Auto Retrovirus 3×10 -3×10 /kg NP Chemo (Cy, Fl) CD28 CD19
Kochenderfer Allo Lentivirus 2.5×105–3.3×109/m2 Chemo Chemo (Cy) CD28, CD20
2012 [29] 4-1BB
Brentjens 2013 Auto Gammaretrovirus 1.5×106-3×106/kg Chemo Chemo (Cy, Mi) CD28 CD19
[30]
Cruz 2013 [31] Allo Retrovirus 1.5×107–1.2×108/m2 SC None CD28 CD19
Kochenderfer Allo Gammaretrovirus 1×106/kg SC None CD28 CD19
2013 [32]
Ritchie 2013 [33] Auto Retrovirus 1.64×109–2.65×109/kg NP Chemo (Cyt, Fl) CD28 LeY
Davila 2014 [4] Auto Retrovirus 3×106/kg NP Chemo (Cy) CD28 CD19
Maude 2014 [23] Auto Lentivirus 7.6×105–2.06×107/kg SC, chemo NP 4-1BB CD19
Wang 2014 [34] Auto Lentivirus 1×107/kg Chemo Chemo (Cy, Ca, De, Et, Vi) 4-1BB LeY
Dai 2015 [35] Allo, Au Lentivirus NP Chemo Chemo (Cy) 4-1BB CD19
Lee 2015 [3] Auto Retrovirus 1×106-3×106/kg SC Chemo (Cy, Fl) CD28 CD19
Porter 2015 [36] Auto Lentivirus 1.4×107–1.1×107/kg Chemo Chemo (Cy, Fl, Pen, Be) 4-1BB CD19
Wang 2015 [37] Auto Lentivirus 1×107/kg Chemo NP 4-1BB CD33
Brudno 2016 [38] Allo Gammaretrovirus 1×106–1×107/kg SC None CD28 CD19
Cai 2016 [39] Auto NP 1.5×107/kg Chemo Chemo 4-1BB CD19
Hu 2016 [40] Auto Retrovirus NP NP Chemo (Cy, Fl) 4-1BB CD19
Kebriaei 2016 Auto Electroporation 1×108/m2 NP Radiation CD28 CD19
[41]
Locke 2016 [42] Auto Retrovirus 2×106/kg SC, biologic, chemo Chemo (Cy or Cy + Fl) CD28 CD19
Prudent 2016 [43] Allo NP NP Chemo, radiation Chemo (Cy, Fl) NP CD19
Turtle 2016a [44] Auto Lentivirus 2×105–2×107/kg SC, chemo Chemo (Cy, Fl) 4-1BB CD19
Turtle 2016b [45] Auto Lentivirus 2×105–2×107/kg SC, chemo Chemo (Cy, Fl) 4-1BB CD19
Wang 2016 [46] Auto Lentivirus 5×107–8×108/kg SC Radiation, biologic (Ri) CD28 CD19
Zhu 2016 [47] Auto Lentivirus 1.19×106–1×106/kg NP Chemo (Cy) 4-1BB CD19
Chen 2017 [48] Allo NP 0.38–4.19×108/kg SC Chemo (Cy, Fl) NP CD19
Fitzgerald 2017 Auto NP NP NP NP 4-1BB CD19
[49]
Fry 2017 [50] Auto Lentivirus 1×106/kg SC, biologic Chemo 4-1BB CD22
Gardner 2017 [51] Auto Lentivirus 0.5×106–1×107/kg SC Chemo (Cy or Cy + Fl) 4-1BB CD19
Hay 2017 [52] Auto Lentivirus 2×105–2×107/kg SC Chemo (Cy, Fl) 4-1BB CD19
Kochenderfer Allo Retrovirus 1-6×106/kg SC Chemo (Cy, Fl) CD28 CD19
2017 [53]
Lai 2017 [54] Auto Lentivirus 5×104/kg NP Chemo (Cy, Fl) CD28 CD19
Neelapu 2017 Auto NP 1.79–5×108/kg SC Chemo CD28 CD19
[55]
Pan 2017 [56] NP Lentivirus 1×105/kg NP Chemo (Cy, Fl) 4-1BB CD19
Qasim 2017 [57] Auto Lentivirus 4×106–4.6×106/kg SC Chemo (Cy, Fl), biologic (Al) CD28, CD19
4-1BB
Ramos 2017 [58] Auto Lentivirus 0.2–2×108/m2 NA NA CD28 CD30
Rossig 2017 [59] Auto Retrovirus 2×108/m2 SC Chemo (Fl, Tr, Th) CD28 CD19
Schuster 2017 Auto Retrovirus 2×108/kg SC, biologic NP 4-1BB CD19
[60]
Smith 2017 [61] NP NP NP NP NP NP CD19
Wang 2017 [62] Auto NP 1.1–2.1×107/kg Chemo Chemo (Cy, Fl) 4-1BB CD30
Zahid 2017 [63] NP NP NP SC, chemo, radiation, Chemo (Cyt, Fl) NP CD19
biologic

Solid malignancies
Morgan 2010 [64] Auto Retrovirus 1×1010 cells Chemo Chemo (Fl, Cy) CD28, ERRB2
4-1BB
Maus 2013 [8] Auto Electroporation 1×108 cells Chemo No lymphodepletion 4-1BB mesothelin
Ahmed 2015 [65] Auto Retrovirus 1×108/m2 Chemo, radiation No lymphodepletion CD28 HER-2
Brown 2015 [66] Auto NP 1×108 cells Chemo, radiation, No lymphodepletion NC IL13-zetakine
biologic
Katz 2015 [67] Auto Retrovirus 1×1010 cells Chemo, radiation No lymphodepletion CD28 CEA
Brown 2016 [68] Auto Lentivirus 1×107 cells Chemo, radiation No lymphodepletion 4-1BB IL13Rα2
Feng 2016 [69] Auto Lentivirus 9.7×106/kg Chemo, radiation Chemo (Cy, Pem, Ci, Do) 4-1BB EGFR
Junghans 2016 Auto Retrovirus 1×1010 cells Chemo, radiation Chemo (Fl, Cy) NC PSMA
[70]
Lamers 2016 [71] Auto Retrovirus 2×109 cells NP No lymphodepletion NC CAIX
You 2016 [72] Auto Lentivirus 5×105/kg NP No lymphodepletion CD28, CAIX
4-1BB
Feng 2017a [73] Auto NP 2.2×106/kg Chemo, radiation No lymphodepletion 4-1BB EGFR
Feng 2017b [74] Auto Lentivirus 2.1×106/kg NP Chemo (Nab-paclitaxel, Cy) 4-1BB HER-2
Hege 2017 [75] Auto Retrovirus 1×108–1010 cells NP No lymphodepleting NP TAG-72
chemotherapy
(continued on next page)
102 E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110

Table 2 (continued)

First author T-cell Transfection/ CAR-T dose Previous treatments Lymphodepletion Co-stim CAR antigen
(year) origin transduction method (cells/m2 or cells/kg) domain target

O'Rourke 2017 Auto Lentivirus 5×108/kg Chemo, radiation, vaccine No lymphodepletion 4-1BB EGFRvIII
[76]
Tanyi 2017 [77] Auto Lentivirus 4.65×107 cells Surgery, chemo, biologic No lymphodepletion 4-1BB mesothelin
Tchou 2017 [78] Auto Electroporation 3×107–3×108 cells NP NP NP c-met
Thistlethwaite Auto Retrovirus 1×109–5×1010 cells Surgery, chemo, Chemo (Fl, Cy) NP CEACAM5
2017 [79] radiation, biologic
Zhang 2017 [80] Auto Lentivirus 1.5×105–1×108/kg Chemo Chemo (Fl or Cy + Fl) CD28 CEA

Abbreviations: Al, alemtuzumab; Allo, allogeneic; Auto, autologous; Be, bendamustine; Ca, carboplatin; Chemo, chemotherapy; Ci, cisplatin; Co-stim, co-stimulatory domain; Cy, cyclophos-
phamide; Cyt, cytarabine; De, dexamethasone; Do, docetaxel; Et, etoposide; Fl, fludarabine; Le, lenalidomide; Mi, mitoxantrone; NC, no costimulatory domains; Pen, pentostatin; Pem,
pemetrexed; Ri, rituximab; SC, stem cell therapy; Tr, treosulpha; Th, thiotepa; Vi, vincristine.

strategy presented in Appendix 2). A peer review of the electronic method of moments approach to investigate potential underlying rela-
search strategy was performed [11]. tionships that may explain some of the variability in response rates. In
addition, we also performed a post hoc analysis by plotting complete re-
Study selection sponse rate against prevalence of cytokine release syndrome to investi-
gate a potential linear relationship. We assessed publication bias using
Search results and full-text articles meeting full eligibility criteria an alternative funnel plot designed for single arm studies [14]. Grading
were reviewed independently and in duplicate (EG, JM) using Distiller of Recommendations, Assessment, Development and Evaluations
Systematic Review Software (DistillerSR, Evidence Partners, Ottawa, (GRADE) was used to assess the quality of the evidence [15].
Canada). Disagreement was settled through discussion with a third re-
viewer (DF, HA, ML, NK). Efficacy outcomes of interest included com- Role of the funding source
plete response (primary outcome), overall or objective response,
relapse, and overall survival. Complete response was defined as the ab- The funder of the study had no role in study design, data collection,
sence of detectable cancer following treatment with CAR-T cell therapy. data analysis, data interpretation, or writing of the report. The corre-
Overall response was defined as the total sum of complete and partial sponding author had full access to all the data in the study and had
responses. Both complete response and overall or objective response final responsibility for the decision to submit for publication.
outcomes excluded patients with continued responses (where patients
were enrolled while in remission from a previous therapy) after receiv- Results
ing CAR-T cell therapy. These patients were included in the other effi-
cacy and safety outcomes (eg, relapse, overall survival). Safety The systematic searches returned 3311 citations. Following
outcomes of interest included cytokine release syndrome, neurotoxic- deduplication and screening by title/abstract and full manuscript, a
ity, infection, and graft versus host disease (GVHD). Patient reported total of 42 hematologic cancer studies (775 patients) and 18 solid can-
outcomes including quality of life and healthy utility measures were cer studies (138 patients) met our full eligibility criteria (Fig 1). Follow-
also extracted. ing screening and de-duplication, there were eight studies that reported
patient data that was fully duplicated [5,16-22] and two studies partially
Data extraction and quality assessment duplicated [23,24].
The characteristics of the included studies are listed in Table 1. All in-
Patient characteristics, CAR-T treatment-specific details, and effi- cluded studies administered CAR-T cell interventions to patients with
cacy/safety outcomes were extracted. Studies were assessed for meth- relapsed or refractory malignancies without control groups. Studies
odological quality using the Institute of Health Economics (IHE) risk of were published between 2008 and 2017, with sample sizes ranging
bias tool [12,13]. We presented in a risk of bias graph using Review from one to 133 participants. 19 (32%) studies investigated acute lym-
Manager 5.3 (London, UK). Data extraction and risk of bias assessments phocytic leukemia (ALL), 18 (30.0%) solid malignancies (eg, liver metas-
were performed independently and in duplicate. Disagreements were tases, glioblastoma), 11 (18%) non-Hodgkin's lymphoma (NHL), two
settled through discussion with a third reviewer (DF, HA, ML, NK). (3%) acute myeloid leukemia, one (2%) chronic lymphocytic leukemia
(CLL), one (2%) Hodgkin's lymphoma and eight (13%) included patients
Data synthesis and analysis with more than one type of hematologic cancer. Thirty-four trials were
reported as phase I studies (57%), 13 were phase I/II (22%), two were
Dichotomous data (eg, complete response) was analyzed and re- phase II studies (3%), and two were reported as being phase 0 (3%).
ported as a proportion with 95% confidence intervals (CI, truncated at Study phase was not reported in 9 studies (15%). Follow-up periods
0 and 1) from the data extracted. Included study results were stratified were highly variable (range: 20 days to 77 months). There was no re-
by CD19 and non-CD19 CAR-T cell constructs for hematologic and solid port of loss to follow-up in any of the included studies. The majority of
malignancies. A random effects model was employed using the studies were performed in either the United States (41 studies, 68%)
DerSimonian and Laird random effects method in order to pool outcome or China (13 studies, 22%).
event rates using Comprehensive Meta-Analysis (version 3; Biostat Inc, The CAR-T cell intervention characteristics are listed in Table 2. A
USA). Statistical heterogeneity between summary data was assessed total of 34 studies (57%) investigated CD19 and 26 (43%) targeted alter-
using the Cochrane I2 statistic. N-of-1 studies were narratively described native tumor associated antigens. There were 49 studies (82%) that in-
and not included in the final meta-analysis. vestigated autologous CAR-T cells, seven (12%) allogeneic, one (2%)
We performed subgroup analyses based on type of cancer, age group mixed, and three (5%) did not report cell type. CAR-T cell therapies
(pediatric and adult), T-cell origin (autologous versus allogeneic), and were defined as autologous regardless of the source of previous stem
construct type (eg, CD19 and non-CD19, co-stimulatory domain cell transplant treatments received (eg, autologous or allogenic). The
types). A post hoc subgroup analysis was performed to assess the effect CAR-T cell dose that was administered to patients varied considerably
of chemotherapeutic preconditioning, as well as the trial phase. A post (2×10 5-1×10 10 cells/kg). There were 25 studies (41.7%) that investi-
hoc random effects meta-regression analysis was performed using the gated CAR-T cells with a 4-1BB co-stimulatory domain, 20 studies
E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110 103

Fig 2. Complete response event rate for patients with hematologic (A-B) and solid malignancies (C). The diamond indicates the pooled summary estimate for each subgroup and the
overall weighted effect estimate where the gray vertical line reflects this pooled summary effect. A random effects model was employed using the DerSimonian and Laird random
effects method.

(33.3%) with a CD28 co-stimulatory domain, seven studies (11.7%) with Complete response was reported in 27 CD19 CAR-T cell therapy he-
no co-stimulatory domain reported, four studies (6.7%) with no co- matologic cancer studies (486 evaluable patients) and seven non-CD19
stimulatory domain and four studies (6.7%) with a CD28 and 4-1BB CAR-T cell therapy studies (65 evaluable patients) as shown in Figure 2A
co-stimulatory domain. and B, respectively. A pooled prevalence of 54.4% [95% CI, 42.5-65.9; I2
We excluded seven solid cancer patients who received surgical re- 69.9%] was demonstrated among patients treated with CD19-targeted
section of their solid tumor prior to CAR-T cell treatment and 30 hema- CAR-T cell therapy. Among the patients that experienced a complete re-
tologic cancer patients who were in continued complete response at the sponse, 52.5% were reported to have achieved absence of minimal resid-
start of CAR-T cell treatment from complete and overall or objective re- ual disease; however it was only reported in three trials (MRD). The
sponse outcomes. sensitivity of the MRD tests performed ranged from 0.01 to 0.08%
The overall risk of bias results from the quality assessment are pre- [39,45,60]. A pooled prevalence of 24.4% [95% CI, 9.4-50.3; I 2 55.8%]
sented in the appendix (Appendix 3). All studies lacked an independent, was demonstrated among patients treated with non-CD19 targeted
blinded assessor and demonstrated high risk of bias with regards to pa- CAR-T cell therapy. Subgroup analysis of CD19+ hematologic cancer
tient recruitment. The majority of studies were also single center trials. types demonstrated that patients with ALL had the highest response
The symmetry observed in the modified funnel plot was not suggestive when compared to CLL and lymphoma. Among the ALL patients, there
of overt publication bias (Appendix 4). was a complete response rate of 77.1% [95% CI, 62.8%-87.1%; I 2 58.5%]
104 E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110

Fig 3. Overall response for patients with hematologic (A-B) and solid tumor malignancies (C). The diamond indicates the pooled summary estimate for each subgroup and the overall
weighted effect estimate where the gray vertical line reflects this pooled summary effect. A random effects model was employed using the DerSimonian and Laird random effects method.

compared to CLL and NHL with rates of 25.5% [95% CI, 13.9%-42.1%; I 2 after controlling for disease type. Patients who received chemother-
0%] and 44.4% [95% CI, 34.1%-55.2%; I2 35.3%], respectively. apeutic preconditioning had significantly higher response rates com-
Among the CD19+ hematologic cancer studies, subgroup analy- pared to those who did not (Appendix 5–10). Meta-regression was
ses of T-cell origin categorized by autologous and allogeneic CAR-T performed among CD19+ hematologic cancer studies to investigate
cell therapies and age subgrouped by pediatric versus adult revealed potential underlying relationships that may explain some of the het-
non-significant differences in complete response. In addition, there erogeneity (Appendix 11). Multiple meta-regression demonstrated
was no difference in response rates observed in our subgroup analy- that ALL patients demonstrated a higher complete response rate
sis by trial phase. In our subgroup analysis of complete response by than CLL and NHL patients after controlling for T-cell origin and co-
co-stimulatory domain type comparing 4-1BB versus CD28, a higher stimulatory domain type (P b .001). Post-hoc analysis using a scatter
response rate was observed with the 4-1BB construct (P b .05) even plot to visually investigate a possible relationship between cytokine
E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110 105

Fig 4. Relapse for patients with hematologic malignancies. The diamond indicates the pooled summary estimate for each subgroup and the overall weighted effect estimate where the gray
vertical line reflects this pooled summary effect. A random effects model was employed using the DerSimonian and Laird random effects method.

release syndrome and complete response demonstrated no relation- treated with CAR-T cell therapy. In post hoc subgroup analyses, the
ship (Appendix 12). prevalence of neurotoxicity did not differ between cancer types, co-
Overall response was reported in 27 CD19 CAR-T cell therapy hemato- stimulatory domains, or trial phase (Appendices 13–15).
logic cancer studies (486 evaluable patients) and seven non-CD19 CAR-T Infection was reported in 14 hematologic cancer studies (272
cell therapy studies (65 evaluable patients) as shown in Figure 3A and B, re- evaluable patients) as shown in Figure 6A. A pooled prevalence of
spectively. A pooled prevalence of 66.5% [95% CI, 56%-75.5%; I2 61.9%] was 12.2% [95% CI, 8.1%-18.0%; I 2 9.9%] was demonstrated among patients
demonstrated among patients treated with CD19 targeted CAR-T cell ther- treated with CAR-T cell therapy.
apy. A pooled prevalence of 47.5% [95% CI, 32.2%-63.2%; I2 23.0%] was dem- Graft versus host disease (GVHD) was reported in three of the seven
onstrated among patients treated with non-CD19 CAR-T cell therapy. hematologic cancer studies that administered an allogeneic product (44
Relapse was reported in 19 CD19 CAR-T cell therapy hematologic evaluable patients) as shown in Figure 6A. A pooled prevalence of GVHD
cancer studies (241 evaluable patients) and five non-CD19 CAR-T cell was 23.4% [95% CI, 8.6%-49.8%; I2 49.7%] was demonstrated among pa-
therapy studies (32 evaluable patients) as shown in Figure 4. A pooled tients treated with CAR-T cell therapy.
prevalence of 37.0% [95% CI, 29.4%-45.4%; I 2 23.7%] was demonstrated B-cell aplasia was measured and reported in seven of the CD19
among patients treated with CD19 targeted CAR-T cell therapy. A CAR-T cell therapy hematologic cancer studies and one non-CD19
pooled prevalence of 75.6% [95% CI, 56.4%-88.2%; I 2 0%] was demon- CAR-T cell therapy hematologic cancer study reviewed. The B-cell
strated among patients treated with non-CD19 CAR-T cell therapy. aplasia persistence ranged from 1 month to 4 years among these
Overall survival was reported in 24 CD19 CAR-T cell therapy hema- studies [3,22,23,26,46,80].
tologic cancer studies (460 evaluable patients) and six non-CD19 CAR- Five N-of-1 hematologic cancer studies were of patients treated with
T cell therapy studies (48 evaluable patients) as shown in Figure 5A CAR-T cell therapy. In four studies that reported on complete response
and B. A pooled analysis was not performed due to the considerable var- and overall response, three patients reported a complete and overall re-
iability in reported follow-up time between trials. Over all CD-19 CAR-T sponse. In three studies that reported on survival to last follow-up, all
cell studies, the survival ranged from 5% to 95%. In non-CD-19 hemato- patients survived. In three studies that reported on cytokine release
logic studies, survival ranged from 74% to 97%. In solid malignancy stud- syndrome, all patients experienced cytokine release syndrome. In
ies, survival ranged from 7% to 92%. three studies that reported on neurotoxicity, all patients experienced
Cytokine release syndrome was reported in 18 hematologic cancer neurotoxic events. In the one study that reported on GVHD, one patient
studies (594 evaluable patients) as shown in Figure 6A. A pooled prev- experienced GVHD. None of the studies reported relapse. Summaries of
alence of 55.3% [95% CI, 40.3%-69.4%; I 2 86.6%] was demonstrated the N-of-1 reports can be found in appendix (Appendix 16).
among patients treated with CAR-T cell therapy. Grading of cytokine Complete response was reported in eight solid cancer studies
release syndrome severity was not consistently reported between (86 evaluable patients) as shown in Figure 2C. A pooled prevalence of
studies. In post hoc subgroup analyses, the prevalence of CRS did not 4.1% [95% CI, 1.6%-10.6%; I 2 0%] was demonstrated among patients
differ between cancer types, co-stimulatory domains, or trial phase treated with non-CD19 CAR-T cell therapy.
(Appendix 13–15). Objective response was reported in ten solid cancer studies (112
Neurotoxicity was reported in 14 hematologic cancer studies (296 evaluable patients) as shown in Figure 3C. The overall objective re-
evaluable patients) as shown in Figure 6A. A pooled prevalence of sponse rate among studies investigating solid tumors was 10.0% [95%
37.2% [95% CI, 28.6%-46.8%; I2 72.6%] was demonstrated among patients CI, 5.1%-18.9%; I2 0%].
106 E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110

Fig 5. Overall survival for patients with hematologic (A-B) and solid malignancies (C). A random effects model was employed using the DerSimonian and Laird random effects method.

Relapse was reported in one solid cancer study (three evaluable reported objective responses, two patients experienced an objective re-
patients). All patients in this study experienced a relapse [66]. sponse. In two studies that reported on cytokine release syndrome, both
Overall survival was reported in nine solid cancer studies (83 patients experienced cytokine release syndrome. In two studies that re-
evaluable patients) as shown in Figure 5C. The overall survival rate ported on survival to last follow-up, both patients experienced mortal-
among studies investigating solid tumors was 34.2% [95% CI, 16.4%- ity. Summaries of N-of-1 reports can be found in the appendix
57.8%; I2 60.6%]. (Appendix 16).
Cytokine release syndrome was reported in two solid cancer studies Patient-reported outcomes, including health-related quality of life,
(17 evaluable patients) as shown in Figure 6B. A pooled prevalence of health utility measures, and patient experience, were not reported in
5.4% [95% CI, 0.8%-30.2%; I 2 0%] was demonstrated among patients any of the reviewed studies.
treated with CAR-T cell therapy. The quality of evidence for the complete response and overall re-
Neurotoxicity was assessed in three solid cancer studies (27 sponse outcomes was assessed as very low due serious risk of bias and
evaluable patients) as shown in Figure 6B. A pooled prevalence of inconsistency (i.e. heterogeneity) in the results (refer to Appendix 17).
12.1% [95% CI, 3.3%-35.7%; I2 3.7%] was determined. This should be interpreted with caution however, as all included trials
Infection was assessed in two solid cancer studies (24 evaluable pa- were single-arm and early-phase in nature.
tients) as shown in Figure 6B. A pooled prevalence of 13.6% [95% CI,
0.7%-79.0%; I 2 72.8%] was demonstrated among patients treated with Discussion
CAR-T cell therapy.
Five N-of-1 solid tumor studies were reports of patients treated with We conducted a systematic review of the efficacy and safety of CAR-
CAR-T cell therapy. In three studies that reported complete response, T cell therapy based on 60 interventional studies that included 913 par-
one patient experienced complete response. In four studies that ticipants. Meta-analysis demonstrated a strong signal of efficacy for
E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110 107

Fig 6. Safety outcomes for patients with hematologic (A) and solid malignancies (B). The diamond indicates the pooled summary estimate for each subgroup and the overall weighted
effect estimate where the gray vertical line reflects this pooled summary effect. A random effects model was employed using the DerSimonian and Laird random effects method.
108 E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110

CD19-targeted CAR-T cell therapy for patients with hematologic malig- Declaration of Interests
nancies. There was significant variability and a lower signal for efficacy
among the non-CD19 targeted CAR-T cell therapy studies. There was no All authors have completed the ICMJE uniform disclosure form at
complete response reported among studies investigating CAR-T cell www.icmje.org/coi_disclosure.pdf and declare: no support from any or-
therapy of solid tumors. ganization for the submitted work; no financial relationships with any
Our review also demonstrated a substantial level of heterogeneity for the organizations that might have an interest in the submitted work in
complete response outcome among the pooled CD19 targeted CAR-T cell the previous three years; no other relationships or activities that could
therapy studies. Meta-regression of the complete response outcome demon- appear to have influenced the submitted work.
strated that cancer type was a significant source of heterogeneity in the
pooled results after controlling for T-cell origin and co-stimulatory domain
Acknowledgments
type. In particular, patients with ALL hematologic cancer had a significantly
higher rate of complete response compared to patients with CLL and NHL.
Biotherapeutics for Cancer Treatment (BioCanRx) supported the
Cytokine release syndrome and neurotoxicity were the most com-
conduct of this study by a grant (Grant reference number: FY17/
monly reported adverse events from CAR-T cell therapy among the he-
CSEI4). BioCanRx is a Government of Canada funded Networks of Cen-
matologic cancer studies. These are unique and often serious side effects
tres of Excellence and was not involved in any other aspect of the pro-
of CAR-T cell therapy, however included studies rarely provided grading
ject, such as the design of the project's protocol and analysis plan, the
of adverse events (which ranged from simple fever, to organ failure),
collection of data and analyses.
which limited further analysis around the severity of side effects. It
ML is supported by The Ottawa Hospital Anesthesia Alternate Funds
has been speculated that the development of cytokine release syndrome
Association and the Scholarship Protected Time Program, Department
is associated with efficacy; however, in our analysis of trial level of data
of Anesthesiology and Pain Medicine, uOttawa.
no association was demonstrated between this adverse event and com-
plete response. This relationship will need to be further explored using
individual patient level data. Novel CAR technologies (eg, universal Funding Source
CARs for multiplexed and logical control of T cell responses) may de-
crease these issues of toxicity. BioCanRx a Government of Canada funded Networks of Centres of
Our review has identified gaps in our current knowledge of CAR-T Excellence.
cell therapy. More complete reporting is needed. For example, MRD is
inconsistently reported, making the level of remission unclear. MRD
Appendix A. Supplementary data
can only be measured in ALL and CLL as there are no routine measures
available in diffuse large B-cell lymphoma or solid tumors, thus limiting
Supplementary data to this article can be found online at https://doi.
our assessment of levels of remission. The outcome ascertainment win-
org/10.1016/j.tmrv.2019.01.005.
dow was also highly variable between studies and frequently limited.
These short follow-up durations limited conclusions on the durability
of response to CAR-T cell therapy. In addition, there was an absence of References
reporting on the safety of CAR-T cell therapy among solid tumor studies,
[1] Jackson HJ, Rafiq S, Brentjens RJ. Driving CAR T-cells forward. Nat Rev Clin Oncol
where only a few studies reported safety outcomes of interest (eg, infec- 2016;13(6).
tion, neurotoxicity). It was also shown in the present review that no [2] Maude S, Barrett DM. Current status of chimeric antigen receptor therapy for hae-
studies investigating CAR-T cell therapy have investigated patient- matological malignancies. Br J Haematol 2016;172(1):11–22.
[3] Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA,
reported outcomes (eg, patient experience, quality of life). Ultimately, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic
this data will be needed to demonstrate that CAR-T cell therapy im- leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet
proves both the duration and quality of life [81]. Finally, the lack of effect 2015;385(9967):517–28.
[4] Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity
in solid tumors suggests that essential issues need to be addressed by
management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia.
research teams (eg, antigen selection) developing future CAR therapies. Sci Transl Med 2014;6(224):224ra225.
This systematic review and meta-analysis provides the largest [5] Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T
cells in chronic lymphoid leukemia. N Engl J Med 2011;365(8):725–33.
synthesis of the clinical results for CAR-T cell therapy in patients with
[6] Yu S, Li A, Liu Q, Li T, Yuan X, Han X, et al. Chimeric antigen receptor T cells: a novel
relapsed or refractory malignancies to date. Despite limitations associ- therapy for solid tumors. J Hematol Oncol 2017;10(1):78.
ated with single arm studies, there is a strong signal of efficacy for [7] Garde D. Juno pulls the plug on a once-promising cancer treatment; 2017.
CD19-targeted CAR-T cell therapy among hematologic malignancies. [8] Maus MV, Haas AR, Beatty GL, Albelda SM, Levine BL, Liu X, et al. T cells expressing
chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res
These results support currently approved treatments, and also identify 2013;1(1):26–31.
knowledge gaps to be addressed by future trials. Our review will help [9] Moher D, Liberati A, Tetzlaff J, Altman DG, Group P. Preferred reporting items for sys-
to better inform patients and other stakeholders of the risks and bene- tematic reviews and meta-analyses: the PRISMA statement. Int J Surg 2010;8(5):
336–41.
fits of CAR-T cell therapy. [10] Grigor EJM, Fergusson DA, Haggar F, Kekre N, Atkins H, Shorr R, et al. Efficacy and
safety of chimeric antigen receptor T-cell (CAR-T) therapy in patients with haema-
Contributors tological and solid malignancies: protocol for a systematic review and meta-
analysis. BMJ Open 2017;7(12):e019321.
[11] McGowan J, Sampson M, Salzwedel DM, Cogo E, Foerster V, Lefebvre C. PRESS peer
Dr. Lalu had full access to all the data in the study and takes full respon- review of electronic search strategies: 2015 guideline statement. J Clin Epidemiol
sibility for the integrity. of the data and the accuracy of the data analysis. 2016;75:40–6.
[12] Guo B, Moga C, Harstall C, Schopflocher D. A principal component analysis is con-
Concept and design: DF, EJMG, ML, NK, HA. ducted for a case series quality appraisal checklist. J Clin Epidemiol 2016;69:
Acquisition of data: EJMG, JM, ML. 199–207.e192.
Analysis or interpretation of data: DF, EJMG, ML, JM, NK, HA, RH. [13] Shuster JJ. Review: Cochrane handbook for systematic reviews for interventions,
version 5.1.0, published 3/2011. Julian P.T. Higgins and Sally green, editors. Res
Drafting of the manuscript: EJMG, ML.
Synth Methods 2011;2(2):126–30.
Critical revision of the manuscript: All authors. [14] Hunter JP, Saratzis A, Sutton AJ, Boucher RH, Sayers RD, Bown MJ. In meta-analyses
Statistical analysis: DF, EJMG, JM. of proportion studies, funnel plots were found to be an inaccurate method of
Obtained funding: DF, ML. assessing publication bias. J Clin Epidemiol 2014;67(8):897–903.
[15] Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. GRADE:
Administrative, technical, or material support: All authors. an emerging consensus on rating quality of evidence and strength of recommenda-
Supervision: DF, ML. tions. BMJ 2008;336(7650):924–6.
E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110 109

[16] Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric an- [41] Kebriaei P, Singh H, Huls MH, Figliola MJ, Bassett R, Olivares S, et al. Phase I trials
tigen receptor–modified T cells for acute lymphoid leukemia. N Engl J Med 2013;368 using sleeping beauty to generate CD19-specific CAR T cells. J Clin Invest 2016;126
(16):1509–18. (9):3363–76.
[17] Zhang Y, Zhang W, Dai H, Wang Y, Shi F, Wang C, et al. An analytical biomarker for [42] Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hosing CM, et al. Phase 1 re-
treatment of patients with recurrent B-ALL after remission induced by infusion of sults of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in re-
anti-CD19 chimeric antigen receptor T (CAR-T) cells. Sci China Life Sci 2016;59(4): fractory aggressive lymphoma. Mol Ther 2017;25(1):285–95.
379–85. [43] Prudent V, Breitbart WS. Chimeric antigen receptor T-cell neuropsychiatric toxicity
[18] Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Feldman Sa, Maric I, et al. Eradi- in acute lymphoblastic leukemia. Palliat Support Care 2017;15(04):499–503.
cation of B-lineage cells and regression of lymphoma in a patient treated with autol- [44] Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. CD19 CAR-T
ogous T cells genetically engineered to recognize Brief report Eradication of B- cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest
lineage cells and regression of lymphoma in a patient treated with autologous T 2016;6:2123–38.
cells. 2010;116(20):4099–102. [45] Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, et al. Immunother-
[19] Kochenderfer JN, Somerville RPT, Lu T, Yang JC, Sherry RM, Feldman SA, et al. Long- apy of non-Hodgkins lymphoma with a defined ratio of CD8+ and CD4+ CD19-
duration complete remissions of diffuse large B cell lymphoma after anti-CD19 chi- specific chimeric antigen receptor-modified T cells. Sci Transl Med 2016;8(355):
meric antigen receptor T cell therapy. Mol Ther 2017;25(10):2245–53. 355ra116.
[20] Saied A, Licata L, Burga RA, Thorn M, McCormack E, Stainken BF, et al. Neutrophil: [46] Wang X, Popplewell LL, Wagner JR, Naranjo A, Blanchard MS, Mott MR, et al. Phase 1
lymphocyte ratios and serum cytokine changes after hepatic artery chimeric antigen studies of central memory – derived CD19 CAR T – cell therapy following autologous
receptor-modified T-cell infusions for liver metastases. Cancer Gene Ther 2014;21 HSCT in patients with B-cell NHL. Blood 2016;127(24):2980–91.
(11):457–62. [47] Zhu Y, Wu Z, Tan Y, Du Y, Liu Z, Ou R, et al. Anti-CD19 chimeric antigen receptor T-
[21] Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, Groot C, cell therapy for adult Philadelphia chromosome-positive acute lymphoblastic leuke-
et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T mia. Medicine 2016;95(51):e5676.
cells: clinical evaluation and management of on-target toxicity. Mol Ther 2013;21 [48] Chen Y, Cheng Y, Suo P, Yan C, Wang Y, Chen Y, et al. Donor-derived CD19-targeted T
(4):904–12. cell infusion induces minimal residual disease-negative remission in relapsed B-cell
[22] Lamers CH, Willemsen R, van Elzakker P, van Steenbergen-Langeveld S, Broertjes M, acute lymphoblastic leukaemia with no response to donor lymphocyte infusions
Oosterwijk-Wakka J, et al. Immune responses to transgene and retroviral vector in after haploidentical haematopoietic stem cell transplantation. Br J Haematol 2017;
patients treated with ex vivo-engineered T cells. Blood 2011;117(1):72–82. 179(4):598–605.
[23] Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen [49] Fitzgerald JC, Weiss SL, Maude SL, Barrett DM, Lacey SF, Melenhorst JJ, et al. Cytokine
receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371(16): release syndrome after chimeric antigen receptor T cell therapy for acute lympho-
1507–17. blastic leukemia. Crit Care Med 2017;45(2):e124–31.
[24] Gardner R, Wu D, Cherian S, Fang M, Hanafi LA, Finney O, et al. Acquisition of a [50] Fry TJ, Shah NN, Orentas RJ, Stetler-Stevenson M, Yuan CM, Ramakrishna S, et al.
CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B- CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to
ALL from CD19 CAR-T-cell therapy. Blood 2016;127(20):2406–10. CD19-targeted CAR immunotherapy. Nat Med 2018;24(1):20–8.
[25] Till BG, Jensen MC, Wang J, Chen EY, Wood BL, Greisman HA, et al. Adoptive immu- [51] Gardner RA, Finney O, Annesley C, Brakke H, Summers C, Leger K, et al. Intent-to-
notherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using treat leukemia remission by CD19 CAR T cells of defined formulation and dose in
genetically modified autologous CD20-specific T cells. Blood 2008;112(6):2261–71. children and young adults. Blood 2017;129(25):3322–31.
[26] Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, et al. T cells with chimeric [52] Hay KA, Hanafi LA, Li D, Gust J, Liles WC, Wurfel MM, et al. Kinetics and biomarkers
antigen receptors have potent antitumor effects and can establish memory in pa- of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified
tients with advanced leukemia. Sci Transl Med 2011;3(95). T-cell therapy. Blood 2017;130(21):2295–306.
[27] Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, Stefanski J, et al. Safety and per- [53] Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, et al. Lymphoma remis-
sistence of adoptively transferred autologous CD19-targeted T cells in patients with sions caused by anti-CD19 chimeric antigen receptor T cells are associated with high
relapsed or chemotherapy refractory B-cell leukemias. Blood 2011;118(18): serum Interleukin-15 levels. J Clin Oncol 2017;35(16):1803–13.
4817–28. [54] Lai Y, Weng J, Wei X, Qin L, Lai P, Zhao R, ... Li P. Toll-like receptor 2 costimulation
[28] Till BG, Jensen MC, Wang J, Qian X, Gopal AK, Maloney DG, et al. CD20-specific adop- potentiates the antitumor efficacy of CAR T cells. Leukemia 2017.
tive immunotherapy for lymphoma using a chimeric antigen receptor with both [55] Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al.
CD28 and 4-1BB domains: pilot clinical trial results. 2015;119(17):3940–51. Axicabtagene Ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N
[29] Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, et al. Bcell Engl J Med 2017;377(26):2531–44.
depletion and remissions of malignancy along with cytokine associated toxicity in a [56] Pan J, Yang JF, Deng BP, Zhao XJ, Zhang X, Lin YH, et al. High efficacy and safety of
clinical trial of antiCD19 chimericantigen receptor–transduced T cells. Blood 2012; low-dose CD19-directed CAR-T cell therapy in 51 refractory or relapsed B acute lym-
119(12):2709–20. phoblastic leukemia patients. Leukemia 2017;31(12):2587–93.
[30] Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, et al. CD19-Targeted T [57] Qasim W, Zhan H, Samarasinghe S, Adams S, Amrolia P, Stafford S, et al. Molecular
Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T
Acute Lymphoblastic Leukemia. Sci Transl Med 2013;5(177):177ra138. cells. Sci Transl Med 2017;9(374):eaaj2013.
[31] Cruz CRY, Micklethwaite KP, Savoldo B, Ramos CA, Lam S, Ku S, et al. Infusion of [58] Ramos CA, Ballard B, Zhang H, Dakhova O, Gee AP, Mei Z, et al. Clinical and immuno-
donor-derived CD19-redirected virus-specific T cells for B-cell malignancies re- logical responses after CD30-specific chimeric antigen receptor-redirected lympho-
lapsed after allogeneic stem cell transplant: a phase 1 study. Blood 2013;122(17): cytes. J Clin Invest 2017;127(9):3462–71.
2965–74. [59] Rossig C, Pule M, Altvater B, Saiagh S, Wright G, Ghorashian S, et al. Vaccination to
[32] Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford W, et al. improve the persistence of CD19CAR gene-modified T cells in relapsed pediatric
Donor-derived anti-CD19 chimeric-antigen-receptor-expressing T cells cause re- acute lymphoblastic leukemia. Leukemia 2017;31(5):1087–95.
gression of malignancy persisting after allogeneic hematopoietic stem cell trans- [60] Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak O, et al. Chimeric antigen
plantation. Blood 2013;122(21):151. receptor T cells in refractory B-cell lymphomas. N Engl J Med 2017;377(26):
[33] Ritchie DS, Neeson PJ, Khot A, Peinert S, Tai T, Tainton K, et al. Persistence and effi- 2545–54.
cacy of second generation CAR T cell against the LeY antigen in acute myeloid leuke- [61] Smith L, Venella K. Cytokine release syndrome: inpatient Care for Side Effects of CAR
mia. Mol Ther 2013;21(11):2122–9. T-cell therapy. Clin J Oncol Nurs 2017;21(2):29–34.
[34] Wang Y, Ying Zhang W, wang Han Q, Liu Y, ren Dai H, lei Guo Y, et al. Effective re- [62] Wang C-M, Wu Z-Q, Wang Y, Guo Y-L, Dai H-R, Wang X-H, et al. Autologous T cells
sponse and delayed toxicities of refractory advanced diffuse large B-cell lymphoma expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin lym-
treated by CD20-directed chimeric antigen receptor-modified T cells. Clin Immunol phoma: an open-label phase I trial. Clin Cancer Res 2017;23(5):1156–66.
2014;155(2):160–75. [63] Zahid U, Shaukat AA, Hassan N, Anwer F. Coccidioidomycosis, immunoglobulin defi-
[35] Dai H, Zhang W, Li X, Han Q, Guo Y, Zhang Y, et al. Tolerance and efficacy of autolo- ciency: safety challenges with CAR T cells therapy for relapsed lymphoma. Immuno-
gous or donor-derived T cells expressing CD19 chimeric antigen receptors in adult therapy 2017;9(13):1061–6.
B-ALL with extramedullary leukemia. OncoImmunology 2015;4(11):1–12. [64] Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report
[36] Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen of a serious adverse event following the administration of T cells transduced with a
receptor T cells persist and induce sustained remissions in relapsed refractory chimeric antigen receptor recognizing ERBB2. Mol Ther 2010;18(4):843–51.
chronic lymphocytic leukemia. Sci Transl Med 2015;7(303):303ra139. [65] Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, et al. Human epi-
[37] Wang Q-s, Wang Y, Lv H-y, Han Q-w, Fan H, Guo B, et al. Treatment of CD33-directed dermal growth factor receptor 2 (HER2) -specific chimeric antigen receptor-
chimeric antigen receptor-modified T cells in one patient with relapsed and refrac- modified T cells for the immunotherapy of HER2-positive sarcoma. J Clin Oncol
tory acute myeloid leukemia. Mol Ther 2015;23(1):184–91. 2015;33(15):1688–96.
[38] Brudno JN, Somerville RPT, Shi V, Rose JJ, Halverson DC, Fowler DH, et al. Allogeneic [66] Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang WC, et al. Bioactivity and
T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B- safety of IL13Ralpha2-redirected chimeric antigen receptor CD8+ T cells in patients
cell malignancies that progress after allogeneic hematopoietic stem-cell transplanta- with recurrent glioblastoma. Clin Cancer Res 2015;21(18):4062–72.
tion without causing graft-versus-host disease. J Clin Oncol 2016;34(10):1112–21. [67] Katz SC, Burga RA, McCormack E, Wang LJ, Mooring W, Point GR, et al. Phase I he-
[39] Cai B, Guo M, Wang Y, Zhang Y, Yang J, Guo Y, et al. Co-infusion of haplo-identical patic immunotherapy for metastases study of intra-arterial chimeric antigen
CD19-chimeric antigen receptor T cells and stem cells achieved full donor engraft- receptor-modified T-cell therapy for CEA+ liver metastases. Clin Cancer Res 2015;
ment in refractory acute lymphoblastic leukemia. J Hematol Oncol 2016;9(1):131. 21(14):3149–59.
[40] Hu Y, Sun J, Wu Z, Yu J, Cui Q, Pu C, et al. Predominant cerebral cytokine release syn- [68] Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, et al. Regression of
drome in CD19-directed chimeric antigen receptor-modified T cell therapy. J glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med 2016;
Hematol Oncol 2016;9(1):70. 375(26):2561–9.
110 E.JM. Grigor et al. / Transfusion Medicine Reviews 33 (2019) 98–110

[69] Feng K, Guo Y, Dai H, Wang Y, Li X, Jia H, et al. Chimeric antigen receptor- [75] Hege KM, Bergsland EK, Fisher GA, Nemunaitis JJ, Warren RS, McArthur JG, et al.
modified T cells for the immunotherapy of patients with EGFR-expressing ad- Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-
vanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci 2016; T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer 2017;5:22.
59(5):468–79. [76] O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD,
[70] Junghans RP, Ma Q, Rathore R, Gomes EM, Bais AJ, Lo AS, et al. Phase I trial of anti- et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates an-
PSMA designer CAR-T cells in prostate Cancer: possible role for interacting interleu- tigen loss and induces adaptive resistance in patients with recurrent glioblastoma.
kin 2-T cell pharmacodynamics as a determinant of clinical response. Prostate 2016; Sci Transl Med 2017;9(399).
76(14):1257–70. [77] Tanyi JL, Stashwick C, Plesa G, Morgan MA, Porter D, Maus MV, et al. Possible compart-
[71] Lamers CH, Klaver Y, Gratama JW, Sleijfer S, Debets R. Treatment of metastatic renal mental cytokine release syndrome in a patient with recurrent ovarian Cancer after
cell carcinoma (mRCC) with CAIX CAR-engineered T-cells-a completed study over- treatment with Mesothelin-targeted CAR-T cells. J Immunother 2017;40(3):104–7.
view. Biochem Soc Trans 2016;44(3):951–9. [78] Tchou J, Zhao Y, Levine BL, Zhang PJ, Davis MM, Melenhorst JJ, et al. Safety and effi-
[72] You F, Jiang L, Zhang B, Lu Q, Zhou Q, Liao X, et al. Phase 1 clinical trial demonstrated cacy of intratumoral injections of chimeric antigen receptor (CAR) T cells in metasta-
that MUC1 positive metastatic seminal vesicle cancer can be effectively eradicated tic breast cancer. Cancer Immunol Res 2017;5(12):1152–61.
by modified anti-MUC1 chimeric antigen receptor transduced T cells. Sci China Life [79] Thistlethwaite FC, Gilham DE, Guest RD, Rothwell DG, Pillai M, Burt DJ, et al. The clin-
Sci 2016;59(4):386–97. ical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T
[73] Feng KC, Guo YL, Liu Y, Dai HR, Wang Y, Lv HY, et al. Cocktail treatment with cells is limited by poor persistence and transient pre-conditioning-dependent respi-
EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells ratory toxicity. Cancer Immunol Immunother 2017;66(11):1425–36.
in a patient with advanced cholangiocarcinoma. J Hematol Oncol 2017;10 [80] Zhang E, Xu H. A new insight in chimeric antigen receptor-engineered T cells for
(1):4. cancer immunotherapy. J Hematol Oncol 2017;10(1):1.
[74] Feng K, Liu Y, Guo Y, Qiu J, Wu Z, Dai H, et al. Phase I study of chimeric antigen re- [81] Hartmann J, Schussler-Lenz M, Bondanza A, Buchholz CJ. Clinical development of
ceptor modified T cells in treating HER2-positive advanced biliary tract cancers CAR T cells-challenges and opportunities in translating innovative treatment con-
and pancreatic cancers. Protein Cell 2018;9(10):838–47. cepts. EMBO Mol Med 2017;9(9):1183–97.

You might also like