• Rates of subsequent malignancies in patients receiving retrovirally modified cellular therapies are similar to those in control patients.

  • Tumors obtained from patients with subsequent malignancies were negative for transgenes and replication competent retrovirus.

Subsequent malignancies are well-documented complications in long-term follow-up of cancer patients. Recently, genetically modified immune effector (IE) cells have shown benefit in hematologic malignancies and are being evaluated in clinical trials for solid tumors. Although the short-term complications of IE cells are well described, there is limited literature summarizing long-term follow-up, including subsequent malignancies. We retrospectively reviewed data from 340 patients treated across 27 investigator-initiated pediatric and adult clinical trials at our center. All patients received IE cells genetically modified with γ-retroviral vectors to treat relapsed and/or refractory hematologic or solid malignancies. In a cumulative 1027 years of long-term follow-up, 13 patients (3.8%) developed another cancer with a total of 16 events (4 hematologic malignancies and 12 solid tumors). The 5-year cumulative incidence of a first subsequent malignancy in the recipients of genetically modified IE cells was 3.6% (95% confidence interval, 1.8% to 6.4%). For 11 of the 16 subsequent tumors, biopsies were available, and no sample was transgene positive by polymerase chain reaction. Replication-competent retrovirus testing of peripheral blood mononuclear cells was negative in the 13 patients with subsequent malignancies tested. Rates of subsequent malignancy were low and comparable to standard chemotherapy. These results suggest that the administration of IE cells genetically modified with γ retroviral vectors does not increase the risk for subsequent malignancy.

Immune effector cells (IECs) genetically modified with γ retroviral vectors (GRVs) or lentiviral vectors expressing chimeric antigen receptor (CAR) T cells are used increasingly to treat patients with hematologic malignancies and are being explored in patients with solid tumors.1-7 When used in combination with lymphodepleting chemotherapy, multiple short-term toxicities of IECs have been reported. However, there is limited literature on potential long-term effects of these therapies, including their genotoxicity.8,9 Subsequent malignancies are among the reported long-term adverse events effects of many standard-of-care cancer therapies, including chemotherapy and hematopoietic stem cell transplant (HSCT), with an incidence of 2% to 5%.8,10,11 Subsequent malignancies remain a potential concern for those receiving GRV-transduced IECs, given reports of their occurrence after transfer of transduced hematopoietic stem cells for inherited diseases such as X-linked severe combined immunodeficiency.12,13 To date, there have been no reported subsequent malignancies in patients receiving GRV-transduced IECs for the treatment of hematologic malignancies or solid tumors.

In this study, we report our Center’s 30-year experience of institutionally generated IECs that were genetically modified using GRVs, representing more than 1000 years of cumulative patient follow-up. The transgenic elements in our studies include components intended to retarget the cells to tumor antigens, improve proliferative capacity, mark the IECs to track persistence, and limit apoptotic activity. We included genetically modified IEC platforms based on activated T cells or virus-specific T lymphocytes (VSTs). We followed both pediatric and adult patients, and a variety of different disease types were included, encompassing both hematologic and solid malignancies. Thirteen patients developed malignancies 1.5 to 172 months after IEC infusion, and we were unable to detect the transferred genetic element in any of the 11 tumors tested. Overall, we observed the same incidence of subsequent cancers in recipients of gene-modified cells as in high-risk patients receiving unmodified IECs.

Study design and logistics

A total of 340 adult and pediatric patients who received ≥1 GRV-transduced genetically modified IECs for a hematologic or solid tumor were retrospectively evaluated on a Baylor College of Medicine (BCM) Institutional Review Board (IRB)-approved protocol. Patients were treated with multiple lines of therapy before enrollment. Their follow-up began on study entry and was conducted by the relevant principal investigator or co-investigator as described in the study-specific calendar. If a patient was lost to follow-up or died on the study, this was recorded at the first available time. If a subsequent malignancy was suspected from clinical symptoms and signs, confirmation was made by laboratory analyses and imaging. Biopsy for transgene testing was requested from all these patients. In our assessment, relapse of the patient’s primary malignancy was not considered a subsequent malignancy. Patients were followed on 28 studies from 1 January 1993 to 4 May 2021.

Five classes of genetically modified IEC were administered to patients: (1) donor-derived gene-marked VSTs in 1 study,14 (2) autologous gene-marked VSTs in 2 studies,15 (3) donor T cells transduced with an inducible caspase 9 suicide gene in 2 studies,16,17 (4) dominant-negative tumor necrosis factor-β receptor–transduced VSTs in 3 studies,18 and (5) CAR T cells with 7 distinct target antigens in 19 studies.19-27 The packaging cell line for donor-marked (ETNA) and automarked (ANGEL/ANGELA) studies used was the PA317 packaging cell line (provided by Genetic Therapy Inc).28 For all other studies, we used the murine embryonic fibroblastic PG13 packaging cell line (obtained from ATCC; #CRL-10686) that produces gibbon ape leukemia virus GalV pseudotyped retroviral particles.29 The retroviral vector backbone for these studies was SFG and (originally provided by R. C. Mulligan, Cambridge, MA).30 Table 1 lists the different constructs used in the study. All patients were treated on protocols approved by the US Food and Drug Administration and by IRBs at St Jude Children’s Research Hospital (3 studies) or BCM (27 studies, including the 3 that were also open at St Jude Children’s Research Hospital). The BCM IRB approved the current retrospective analysis.

Table 1.

Different types of immune effectors included in the study

Neo marker geneTGF-β DNRCAR/cytokineCARiC9
Auto-VSTs    
Donor VSTs   
Donor allodepleted ATCs     
Auto ATCs   
PA317*     
PG13  
Neo marker geneTGF-β DNRCAR/cytokineCARiC9
Auto-VSTs    
Donor VSTs   
Donor allodepleted ATCs     
Auto ATCs   
PA317*     
PG13  

allo, allogeneic; ATC, activated T cell; auto, autologous; DNR, dominant-negative receptor; iC9, inducible caspase 9.

*

PA317: amphitropic retrovirus packaging cell line was used for auto-VSTs and donor VSTs (obtained by Genetic Therapy Inc).

PG13: murine embryonic fibroblastic PG13 packaging cell lines (obtained from ATCC; #CRL-10686) were used for all other GRV-IEC types.

To determine whether GRV genetic modification of IECs increases the risk of subsequent malignancies, we included a control group of patients who received IECs that had not been genetically modified before infusion. This group included 111 patients with Hodgkin lymphoma (HL) or B-cell non-Hodgkin lymphoma (NHL) who were treated with Epstein-Barr VSTs (EBVSTs). Like the genetically modified cell group, patients were evaluated for time to second malignancy starting at enrollment.

GRV transgene and replication-competent retrovirus testing

GRV transgenes were detected via quantitative polymerase chain reaction (PCR) amplification of peripheral blood mononuclear cells (PBMCs) collected preinfusion and at standardized times after IEC infusion as per the study calendar. Transgene copy number in the infused cell lines and the follow-up blood samples of patients on protocols with genetically modified T cells are measured by quantitative PCR (qPCR), using TaqMan primers and probes custom designed to detect the specific transgene in genomic DNA extracted from the IEC product and patients’ PBMC as previously described for the individual studies.14-28 The National Gene Vector Biorepository at Indiana University (Indianapolis, IN) tested samples for replication competent retrovirus (RCR) in PBMCs preinfusion and subsequently as required by the study-specific calendar.

Outcome and statistical design

Descriptive statistics were used to summarize patient characteristics.31 Cumulative follow-up was used to describe the total duration of follow-up for all patients on the treatment group by summing all the individual follow-up years of each patient. The primary endpoint of this study was the presence of a subsequent malignancy. Time to subsequent malignancy was recorded from the time of the first infusion on the study to the time of the first diagnosis of subsequent disease. The cumulative incidence of these malignancies was estimated and compared between genetically modified IECs and genetically unmodified modified IECs by the competing risk method as described by Gray.32 The subdistribution hazard ratios were estimated and compared between the treatment groups using the Fine-Gray subdistribution hazard regression model.33 Rates are calculated with exact 95% binomial confidence intervals (CIs). All P values are 2-sided, and values of P < .05 were considered statistically significant.34 

Patient characteristics

A total of 340 patients received GRV genetically modified IECs. Of these, 204 (60.0%) were adult and 136 (40.08%) were pediatric patients with a median age at enrollment of 23.5 years (range, 1-78). One hundred and eighty-eight (55.3%) patients had primary hematologic malignancy, 150 (44.1%) had solid tumors, and 2 (0.6%) had nonmalignant conditions. The most common hematologic malignancy was NHL, followed by HL and acute lymphoblastic leukemia. Among solid tumors, osteosarcoma and neuroblastoma were the most common (Table 2). Of the 340 patients, 99 (29.1%) had autologous HSCT and 83 (24.4%) had an allogeneic HSCT. The median follow-up of patients was 14.9 months (range, >1-183), with 192 (56.5%), 72 (21.2%), and 29 (8.5%) patients followed for more than 1, 5, and 10 years, respectively.

Table 2.

Baseline patient characteristics

CharacteristicAll patients, n = 340
Age, median (range), y 23.5 (1-78) 
Sex, n (%)  
 Male 197 (57.9) 
 Female 143 (42.1) 
Primary diagnosis, n (%)  
 Hematologic malignancy 188 (55.3) 
 Solid malignancy 150 (44.1) 
 Nonmalignant conditions 2 (0.6) 
Hematologic malignancies, n (%)  
 ALL 46 (24.5) 
 AML 11 (5.9) 
 CLL 11 (5.9) 
 CML 8 (4.3) 
 NHL 41 (21.8) 
 HL 54 (28.7) 
 MM 7 (3.7) 
 Others 10 (5.3) 
Solid tumors, n (%)  
 Neuroblastoma 33 (22.0) 
 Glioblastoma 19 (12.7) 
 Lung cancer 10 (6.7) 
 Osteosarcoma 52 (34.7) 
 Head and neck 16 (10.7) 
 Liver tumors 8 (5.3) 
 Others 12 (8.0) 
Transplant, n (%)  
 Autologous 99 (29.1) 
 Allogeneic 83 (24.4) 
 No transplant 158 (46.5) 
History of radiation therapy, n = 339 (%)  
 No 137 (40.4) 
 Yes 202 (59.6) 
 Median follow-up (range), mo 14.9 (<1-183) 
 Cumulative follow-up, y 1027 
CharacteristicAll patients, n = 340
Age, median (range), y 23.5 (1-78) 
Sex, n (%)  
 Male 197 (57.9) 
 Female 143 (42.1) 
Primary diagnosis, n (%)  
 Hematologic malignancy 188 (55.3) 
 Solid malignancy 150 (44.1) 
 Nonmalignant conditions 2 (0.6) 
Hematologic malignancies, n (%)  
 ALL 46 (24.5) 
 AML 11 (5.9) 
 CLL 11 (5.9) 
 CML 8 (4.3) 
 NHL 41 (21.8) 
 HL 54 (28.7) 
 MM 7 (3.7) 
 Others 10 (5.3) 
Solid tumors, n (%)  
 Neuroblastoma 33 (22.0) 
 Glioblastoma 19 (12.7) 
 Lung cancer 10 (6.7) 
 Osteosarcoma 52 (34.7) 
 Head and neck 16 (10.7) 
 Liver tumors 8 (5.3) 
 Others 12 (8.0) 
Transplant, n (%)  
 Autologous 99 (29.1) 
 Allogeneic 83 (24.4) 
 No transplant 158 (46.5) 
History of radiation therapy, n = 339 (%)  
 No 137 (40.4) 
 Yes 202 (59.6) 
 Median follow-up (range), mo 14.9 (<1-183) 
 Cumulative follow-up, y 1027 

ALL, acute lymphocytic leukemia; AML, acute myelocytic leukemia; CML, chronic myelocytic leukemia; MM, multiple myeloma; TBI, total body irradiation.

The control group consisted of 111 patients treated with genetically unmodified EBVSTs. Their median age was 44 (range, 6-78). All patients had lymphoma, 51 (45.9%) with HL and 60 (54.1%) with B-cell NHL. Of the 111 patients, 37 (33.3%) had autologous HSCT and 13 (11.7%) had an allogeneic HSCT (Table 3).

Table 3.

Control group baseline characteristics

CharacteristicAll patients, n = 111
Age, median (range), y 44 (6-78) 
Sex, n (%)  
 Male 73 (65.8) 
 Female 38 (34.2) 
Primary diagnosis (%)  
 Hodgkin lymphoma 51 (45.9) 
 Non-Hodgkin lymphoma 60 (54.1) 
Transplant, n (%)  
 Autologous 37 (33.3) 
 Allogeneic 13 (11.7) 
 No transplant 61 (55.0) 
 Included protocols GRALE (NCT01555892)
ALCI and ALASCER (NCT00062868) 
CharacteristicAll patients, n = 111
Age, median (range), y 44 (6-78) 
Sex, n (%)  
 Male 73 (65.8) 
 Female 38 (34.2) 
Primary diagnosis (%)  
 Hodgkin lymphoma 51 (45.9) 
 Non-Hodgkin lymphoma 60 (54.1) 
Transplant, n (%)  
 Autologous 37 (33.3) 
 Allogeneic 13 (11.7) 
 No transplant 61 (55.0) 
 Included protocols GRALE (NCT01555892)
ALCI and ALASCER (NCT00062868) 

General characteristics in patients with subsequent malignancies

Of the 340 patients treated with a cumulative follow-up of 1027 years, 13 (3.8%; 95% CI, 2.1-6.4) patients developed a total of 16 subsequent malignancies (Table 4). Three of the 13 (23%) patients had more than 1 subsequent malignancy. The median age of all patients with subsequent malignancies at enrollment was 27 (range, 6-62). Ten of the 13 patients were males. Twelve of 16 (75%) subsequent malignancies were solid tumors, the most common being basal cell carcinoma, with 4/16 (25%) total malignancies. There was a broad range of tumors diagnosed within this solid tumor subgroup with no site favored (Table 4). Subsequent malignancies occurred 1.5 to 172 months (median, 45 months) after IEC infusion, with 10 (62.5%) malignancies occurring within 5 years and 6 (38%) malignancies occurring more than 10 years after IEC therapy. Of the 13 patients, 8 were still alive at the last follow-up. Subsequent malignancy contributed to death in 2 patients, although 5 died with subsequent malignancies.

Table 4.

Characteristics of patients that developed subsequent malignancies after genetically modified IECs

PatientAge at enrollment (y)SexPrimary diseaseHSCTIEC typeSubsequent neoplasmTime from IEC therapy,
mo
Tissue transgene detectionRCRTime to LFU,
y
Outcome at last follow-up
Neuroblastoma Yes
(auto-HSCT) 
GD2 CAR T AML 1.5 Negative Negative Alive 
27 HL Yes
(auto-HSCT) 
DNR TGF-β EBVSTs MDS 24 Negative Negative Died of complications after allogeneic transplant 
17 Nasopharyngeal carcinoma No DNR TGF-β EBVSTs Spindle-cell neoplasm 24 Negative Negative 4.5 Alive 
62 CLL Yes
(allo-HSCT) 
CD19 CAR T Bladder cancer 48 Negative Negative Died of secondary infection 
11 Glioblastoma No HER2 CAR T T-cell lymphoma 36 Negative Negative Died of T-cell lymphoma 
60 CLL No CD19 CAR T BCCx2* 36 and 48 Unable to obtain Negative Alive 
47 HL Yes
(auto-HSCT) 
DNR TGF-β EBVSTs Breast ductal cell carcinoma 42 Negative Negative Alive 
55 NHL Yes
(auto-HSCT) 
CD19 CAR T BCC 36 Unable to obtain Negative 7.5 Alive 
56 NHL No CD19 and κ CAR T 1-Penile squamous cell carcinoma 72 Negative Negative 7.5 Died of MDS complications 
2-MDS 36 Negative Negative 
10 36 HL Yes
(auto-HSCT) 
Auto EBVSTs with NeoR 1-RCC 154 Unable to obtain Negative 15 Alive 
2-Meningioma 172 Unable to obtain Unable to obtain 
11 18 ALL and secondary AML Yes
(allo-HSCT) 
Allo EBVSTs with NeoR Nerve sheath tumor 96 Negative Negative 15 Died of unrelated accident 
12 AML Yes
(allo-HSCT) 
Allo EBVSTs with NeoR Thyroid adenoma 144 Unable to obtain Negative 15 Alive 
13 15 ALL Yes
(allo-HSCT) 
Allo EBVSTs with NeoR BCC 156 Negative Negative 15 Alive 
PatientAge at enrollment (y)SexPrimary diseaseHSCTIEC typeSubsequent neoplasmTime from IEC therapy,
mo
Tissue transgene detectionRCRTime to LFU,
y
Outcome at last follow-up
Neuroblastoma Yes
(auto-HSCT) 
GD2 CAR T AML 1.5 Negative Negative Alive 
27 HL Yes
(auto-HSCT) 
DNR TGF-β EBVSTs MDS 24 Negative Negative Died of complications after allogeneic transplant 
17 Nasopharyngeal carcinoma No DNR TGF-β EBVSTs Spindle-cell neoplasm 24 Negative Negative 4.5 Alive 
62 CLL Yes
(allo-HSCT) 
CD19 CAR T Bladder cancer 48 Negative Negative Died of secondary infection 
11 Glioblastoma No HER2 CAR T T-cell lymphoma 36 Negative Negative Died of T-cell lymphoma 
60 CLL No CD19 CAR T BCCx2* 36 and 48 Unable to obtain Negative Alive 
47 HL Yes
(auto-HSCT) 
DNR TGF-β EBVSTs Breast ductal cell carcinoma 42 Negative Negative Alive 
55 NHL Yes
(auto-HSCT) 
CD19 CAR T BCC 36 Unable to obtain Negative 7.5 Alive 
56 NHL No CD19 and κ CAR T 1-Penile squamous cell carcinoma 72 Negative Negative 7.5 Died of MDS complications 
2-MDS 36 Negative Negative 
10 36 HL Yes
(auto-HSCT) 
Auto EBVSTs with NeoR 1-RCC 154 Unable to obtain Negative 15 Alive 
2-Meningioma 172 Unable to obtain Unable to obtain 
11 18 ALL and secondary AML Yes
(allo-HSCT) 
Allo EBVSTs with NeoR Nerve sheath tumor 96 Negative Negative 15 Died of unrelated accident 
12 AML Yes
(allo-HSCT) 
Allo EBVSTs with NeoR Thyroid adenoma 144 Unable to obtain Negative 15 Alive 
13 15 ALL Yes
(allo-HSCT) 
Allo EBVSTs with NeoR BCC 156 Negative Negative 15 Alive 

ALL, acute lymphocytic leukemia; Allo, allogeneic; AML, acute myelocytic leukemia; auto, autologous; BCC, basal cell carcinoma; CLL, chronic lymphoid leukemia; LFU, last follow-up; MDS, myelodysplastic syndrome; NeoR, neomycin resistant marker gene; RCC, renal cell carcinoma; RCR, replication-competent retrovirus; TGF, transforming growth factor.

*

Patient was diagnosed with 2 basal cell carcinomas of the skin at 36 and 48 mo after infusion.

Patient received both CD-19 and κ CAR. Time to last follow-up and to secondary malignancy was calculated from time of his first infusion (CD19 CAR).

Patient with history of neurofibromatosis.

Cumulative incidence of subsequent malignancies in gene-modified IEC recipients

We compared the cumulative incidence of the first subsequent malignancy among patients receiving genetically modified IECs group vs patients treated with genetically unmodified EBVSTs. Of 340 patients treated, 13 (3.8%) developed subsequent malignancies. A total of 195 (57.4%) died and 132 (38.8%) were alive at the last follow-up (up to 15.2 years) without subsequent malignancies. We observed that the 5-year cumulative incidence of the first subsequent malignancy in the recipients of genetically modified cells was 3.6% (95% CI, 1.8-6.4). Of the 111 patients receiving genetically unmodified cells, 4 patients (3.6%) developed subsequent malignancies (2 myelodysplastic syndrome, 2 sarcoma) at 5 years, whereas 39 (35.1%) died and 68 (61.3%) were alive at the last follow-up (up to 5 years) without subsequent malignancies. The 5-year cumulative incidence of subsequent malignancy in this population was 4.2% (95% CI, 1.4-9.7) (Figure 1). Thus, at 5 years, there was no difference in the cumulative incidence of the first subsequent malignancy between the 2 groups (P = .953, Gray test). The estimated subdistribution hazard ratio of modified cells compared with unmodified was 0.9 (95% CI, 0.30-2.9), and thus there was no significant effect for the cumulative incidence of subsequent malignancies (P = .859 by the Fine-Gray subdistribution hazard regression model).

Figure 1.

Cumulative incidence of subsequent malignancies. Cumulative incidence plots showing incidence of γ retroviral genetically modified IECs vs genetically unmodified IECs.

Figure 1.

Cumulative incidence of subsequent malignancies. Cumulative incidence plots showing incidence of γ retroviral genetically modified IECs vs genetically unmodified IECs.

Close modal

Distribution of subsequent malignancies across studies with genetic modification

Neither the source of IEC nor the transgene itself discernibly influenced the risk of subsequent malignancies. Of the 16 subsequent malignancies reported, 3 occurred in patients receiving donor gene-marked IECs, 2 in recipients of autologous gene-marked IECs, 3 in recipients of IECs expressing a dominant negative receptor for tumor necrosis factor-β, and 8 in patients treated with CAR T cells. Patients on 3 additional CAR T-cell studies had no subsequent malignancies (CD30, glypican 3, and CD5); although the CD30 study opened 10 years ago, the glypican-3 and CD5 studies started enrolling patients in the past 5 years (Table 5).

Table 5.

Details of included clinical trials

Type of modificationType of immune effector cellsTrial names (cumulative follow-up, y)Trials included (NCI no.)No. of patientsNo. of SMsMedian time to last detected transgene, mo (range)*
Donor marked EBVSTs ETNA (287.6) NCT00058812 26 44.5 (0-105) 
Auto marked EBVSTs ANGEL and ANGELA (19.1) NCT00058617 Up to 2 y 
iCasp9 iCasp9 T cell CASPALLO (48.3)
DOTTI (28.0) 
NCT00710892
NCT01494103 
24 12 (0.25-108) 
Dominant negative receptor DNR TGF-β cells TGFB (38.1)
RESIST NPC (17.9)
HESTIA (13.3) 
NCT00675571
NCT02065362
NCT02379520 
28 6 (0.5-72) 
CAR T cell GD2 CAR T cells NESTLES (93.1)
GRAIN (30.0)
GAIL-N (10.3)
VEGAS (16.2) 
NCT00085930
NCT01822652
NCT03635632
NCT01953900
NCT03294954 
51 3 (0-108) 
CD19 CAR T cells CRETI (59.2)
ATECRAB (8.5)
SAGAN (47.0)
CARSPASCIO (15.5)
MULTIPRAT (55.6) 
NCT00586391
NCT00709033
NCT01853631
NCT02050347
NCT00840853 
63 6 (0-96) 
HER2 CAR T cells HERCREEM (20.6)
HERT-GBM (15.9)
HEROS (71.1) 
NCT00889954
NCT01109095
NCT00902044 
71 1.5 (0-84) 
CD30 CAR T cells CAR CD30 (18.6)
CART CD30 (30.3)
RELY 30 (26.0) 
NCT01192464
NCT01316146
NCT02917083 
33 6 (0-24) 
Glypican 3 CAR T cells GLYCAR (1.8)
GAP (3.6) 
NCT02905188
NCT02932956 
3 (2-12) 
CD5 CAR T cells MAGENTA (6.7) NCT03081910 11 3 (0.75-30) 
κ CAR T cells CHARKALL (45.1) NCT00881920 18 2 6 (0.25-60) 
Type of modificationType of immune effector cellsTrial names (cumulative follow-up, y)Trials included (NCI no.)No. of patientsNo. of SMsMedian time to last detected transgene, mo (range)*
Donor marked EBVSTs ETNA (287.6) NCT00058812 26 44.5 (0-105) 
Auto marked EBVSTs ANGEL and ANGELA (19.1) NCT00058617 Up to 2 y 
iCasp9 iCasp9 T cell CASPALLO (48.3)
DOTTI (28.0) 
NCT00710892
NCT01494103 
24 12 (0.25-108) 
Dominant negative receptor DNR TGF-β cells TGFB (38.1)
RESIST NPC (17.9)
HESTIA (13.3) 
NCT00675571
NCT02065362
NCT02379520 
28 6 (0.5-72) 
CAR T cell GD2 CAR T cells NESTLES (93.1)
GRAIN (30.0)
GAIL-N (10.3)
VEGAS (16.2) 
NCT00085930
NCT01822652
NCT03635632
NCT01953900
NCT03294954 
51 3 (0-108) 
CD19 CAR T cells CRETI (59.2)
ATECRAB (8.5)
SAGAN (47.0)
CARSPASCIO (15.5)
MULTIPRAT (55.6) 
NCT00586391
NCT00709033
NCT01853631
NCT02050347
NCT00840853 
63 6 (0-96) 
HER2 CAR T cells HERCREEM (20.6)
HERT-GBM (15.9)
HEROS (71.1) 
NCT00889954
NCT01109095
NCT00902044 
71 1.5 (0-84) 
CD30 CAR T cells CAR CD30 (18.6)
CART CD30 (30.3)
RELY 30 (26.0) 
NCT01192464
NCT01316146
NCT02917083 
33 6 (0-24) 
Glypican 3 CAR T cells GLYCAR (1.8)
GAP (3.6) 
NCT02905188
NCT02932956 
3 (2-12) 
CD5 CAR T cells MAGENTA (6.7) NCT03081910 11 3 (0.75-30) 
κ CAR T cells CHARKALL (45.1) NCT00881920 18 2 6 (0.25-60) 

DNR, dominant-negative receptor; NCI, National Cancer Institute; TGF, transforming growth factor.

*

Time to last detected transgene was calculated from the time of last infusion (if patient received >1 infusion) to time to any positive transgene value detected by qPCR.

Two subsequent malignancies were reported in a patient who received both CD19 and κ CAR T cells.

Transgene detection and RCR testing

To determine whether gene-modified cells directly contributed to subsequent malignancies, we used qPCR to measure transgene prevalence in peripheral blood and tumors. All 13 subsequent malignancy patients had RCR testing of peripheral blood at multiple time points after IEC therapy and no patient had RCR detectable in PBMCs before or after malignancy onset. Table 5 includes IEC and study-specific details on transgene persistence. Additionally, 11 of 13 patients with subsequent malignancy had tumor biopsies available. We did not detect any IEC transgenes in any of these 11 biopsies.

Although 1 patient developed T-cell lymphoma 2 years following HER2–CAR T-cell therapy, he was previously diagnosed with constitutional mismatch repair deficiency syndrome, an aggressive cancer predisposition syndrome with mediastinal T-cell lymphoma being the most common.35,36 No IEC transgene was detected in the lymphoma biopsy, and RCR testing was negative at all time points before and after infusion.

We reviewed 340 patients who received ≥1 GRV-modified IEC with a cumulative follow-up of 1027 years. To do date, this is the longest follow-up of patients receiving retroviral genetically modified IECs. Our cohort of patients included a wide range of ages (range, 1-78 years), as well as varied primary diagnoses, encompassing solid tumors, malignant hematologic diseases, and nonmalignant immunodeficiencies. In this retrospective analysis, we demonstrate no increased risk of subsequent malignancy in patients treated with retroviral genetically modified IECs. Additionally, all patients treated on this study had exposure to chemotherapy before enrollment on each respective clinical trial. We show that the risk of subsequent malignancy in this patient group is 3.6%, which is similar to the 2% to 5% range that has been documented in previous literature for patients treated with chemotherapy.7 

In this study, RCR testing was negative in all patients at all time points, and no IEC transgenes were detected in any tumor biopsies form patients with subsequent malignancies after IEC infusion. These findings reinforce Bear et al’s study on RCR detection, in which they looked at 27 clinical studies using genetically modified T-cell products with GRVs and did not detect any RCR in 42 viral supernatant lots or any T-cell products and Cornetta et al’s evaluation of 241 patients over 14 clinical trials, none of whom had detectable RCR infection.37,38

Although retroviral genetically modified IECs do not appear to increase the risk of subsequent malignancy in our study, other factors which contribute to malignant transformation and necessitate close follow-up. For example, Brown et al examined whether CD40 ligand (CD40L) deficiency, known as X-linked hyper-IgM syndrome, could be corrected via gene therapy. The authors transduced murine bone marrow or thymic cells with a retroviral vector containing complementary DNA for murine CD40 ligand and injected them into deficient mice. Although treatment led to improved immune response in mice, 12 of 19 mice developed T-cell lymphomas. Their data suggested that retroviral genetic modification did not lead to malignant transformation because RCR was not detected in samples, nor were retroviral particles identified by electron microscopic examination in tumors. Furthermore, insertional mutagenesis seemed unlikely because proviral DNA was only noted in a small group of tumor cells. Instead, subsequent malignancies were thought to be secondary to CD40L’s role in thymocyte regulation via upregulation of costimulatory molecules on thymocyte accessory cells and naïve T lymphocytes. Thus, the constitutive expression of CD40L in immature thymocytes potentially lead to a malignant T-cell clone because the CD40L transgene was detected in tumor tissue in these mice.39 

Although no lentiviral genetically modified IECs have been associated with secondary malignancies to date, clonal expansion from insertional mutagenesis and subsequent malignancies remain a risk. Shah et al reported 1 patient presenting with asymptomatic leukocytosis around 50 days after receiving treatment with lentiviral genetically modified CD22-CAR T cells for relapsed B-cell ALL. A dominant T-cell clone containing a copy of the lentiviral vector integrated into the intron of the CBL gene was detected. Following treatment with steroids the patient was reevaluated at day +180, and the clone was not detected.40 Similarly, Fraietta et al reported a case of clonal CAR T-cell expansion in the context of anti-CD19 CAR T-cell therapy for a 70 year old with relapsed chronic lymphocytic leukemia. This was likely because of a concomitant lentiviral integration into the TET2 gene, along with a hypomorphic mutation on the patient’s second allele. Because TET2 plays a role in regulating hematopoiesis and T-cell differentiation, this integration likely led to the patient’s CAR T-cell expansion and antitumor effect.41 These cases of clonal expansion demonstrate that lentiviral integration into host genes pose a risk of insertional mutagenesis, prompting a need for continued long-term follow-up.

Other genetic transfer systems, such as the piggyBac transposon system, offer significant advantages to viral vectors because of lower cost and ability to transfer large genetic inserts42,43; however, a theoretical risk of genomic rearrangement and malignant transformation remains. Bishop et al used CD19-CAR T cells generated with the piggyBac transposon system to treat 10 CD19+ leukemia and lymphoma patients. Although treatment appeared effective, 2 of 10 patients developed secondary monoclonal CAR T-cell malignancies 12 months and 16 months after treatment. CAR T expression was noted in biopsies of both patients, indicating malignant transformation; neither malignancy had integration of the CAR transgene into oncogenes, and there was no unique driver of malignant transformation for the 2 patients. Bishop et al note that their production protocol, which included electroporation and varying concentrations of transposon and transposase, were likely factors in the malignant transformation of these patients.44 Of note, Gregory et al recently treated 23 patients using a piggyBac transposon generated CAR T cells targeting B-cell maturation antigen. There were no significant toxicities noted, nor were there subsequent malignancies at 3 years posttherapy.45,46

Additionally, gene-editing strategies including CRISPR-Cas9 and transcription activator-like effector nucleases have been used to modify CAR T cells to reduce the risk of graft-versus-host disease or rejection.47 Transcription activator-like effector nuclease disruption of the TRAC gene and the CD52 locus was recently associated with a chromosomal abnormality on chromosome 14, the location of the TRAC gene, but in a region associated with myeloproliferative disorders. Although there was clonal expansion of the modified cells, no progression to malignancy has occurred to date.48 

In an initial analysis of 48 of our patients receiving GRV-modified cells, we did not see an association between malignant transformation and vector copy number. We observed a mean vector copy number per transduced cell of 5.38 (range, 1.48-9.40). That said, most studies for which vector copy number was calculated are within the first 5 years of follow-up; thus, more long-term assessment is necessary.44 

The 1 patient on our study who developed a mediastinal T-cell lymphoma after HER2-CAR T-cell infusion had no evidence of the CAR T transgene in the tumor biopsy sample nor in the peripheral blood. Additionally, he was known to have constitutional mismatch repair deficiency, which put him at increased risk for hematologic malignancies, with mediastinal lymphoma being particularly prevalent in patients with this deficiency. There is no evidence to date of subsequent resulting from GRV modification in T cells, as demonstrated by multiple groups.49,50

In summary, our data reinforce the safety of retroviral genetically modified IECs, and in our series show that GRV-modified vectors did not significantly increase the risk of subsequent malignancy in patients. Additional long-term studies will be needed to further demonstrate the safety and risk of subsequent malignancies in the transposon systems and in lentiviral IECs.

The authors thank all the physicians who cared for these patients, the Good Manufacturing Practice (GMP) facility, Good Laboratory Practice (GLP) facility, and clinical research staff who participated in these studies and the patients for participating in long-term follow up.

This work was supported by grants from the National Institutes of Health, National Cancer Institute (P50CA126752, 1U54CA232568-01, and P01CA094237), Stand Up To Cancer (SU2C)/St Baldrick’s Pediatric Cancer Dream Team Translational Research Grant (SU2C-AACR-DT1113), Stand Up To Cancer (SU2C)/American Association for Cancer Research (AACR) 604817 Meg Vosburg T-Cell Lymphoma Dream Team, and the Leukemia and Lymphoma Society. SU2C is a program of the Entertainment Industry Foundation administered by the AACR.

Contribution: D.H.M.S. and I.N.M. drafted the manuscript; T.W. and M.W. performed statistical analyses; H.E.H., C.A.R., L.C.H., D.H.M.S., and I.N.M. designed the study; D.H.M.S. and I.N.M. performed data collection; N.A., M.H., S.B., O.D., S.G., S.B.W., P.D.L., M.M., B.O., R.H.R., A.H., L.S.M., L.H., C.A.R., C.M.R., M.K.B., and H.E.H. contributed to products manufacturing, samples analysis, patient enrollment, and edited the manuscript; and all authors approved the final version of the manuscript.

M.K.B. and C.M.R. have equity in Allovir, Marker Therapeutics, and Tessa Therapeutics; have served on advisory boards for Walking Fish Therapeutics, CellGenix GmbH, Marker Therapeutics, Tessa Therapeutics, Abintus, Allogene, Bellicum Pharmaceuticals, Bluebird Bio, Athenex, Memgen, Turnstone Biologics, Coya Therapeutics, TScan Therapeutics, Onkimmune, Poseida Therapeutics, and Allovir; and have received research support from Tessa Therapeutics. H.E.H. has equity in Allovir and Marker Therapeutics; has served on advisory boards for Tessa Therapeutics, Novartis, Gilead, GSK, Kiadis, and Fresh Wind Biotechnologies; and received research support from Tessa Therapeutics and Kuur Therapeutics. S.G. has a consulting agreement with Tessa Therapeutics; is a compensated DSMB member of Immatics; has received honoraria from Tidal, Catamaran Bio, and Novartis within the last 2 years; and has received research support from Tessa Therapeutics in the past. P.L. served on an advisory board for Karyopharm Therapeutics. B.G. owns QBRegulatory Consulting, which has agreements with LOKON Pharma, TESSA Therapeutics, Marker Therapeutics, and AlloVir. L.M. and A.H. report research support from Athenex, Inc.

Correspondence: David Steffin, Center for Cell and Gene Therapy, Baylor College of Medicine/Texas Children's Hospital, Feigin Center, 1102 Bates St, Ste 730.05, Houston, TX 77030; e-mail: dhsteffi@texaschildrens.org.

There is a Blood Commentary on this article in this issue.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

1.
June
CH
,
Sadelain
M
.
Chimeric antigen receptor therapy
.
N Engl J Med.
2018
;
379
(
1
):
64
-
73
.
2.
Klebanoff
CA
,
Rosenberg
SA
,
Restifo
NP
.
Prospects for gene-engineered T cell immunotherapy for solid cancers
.
Nat Med.
2016
;
22
(
1
):
26
-
36
.
3.
Sadelain
M
,
Rivière
I
,
Riddell
S
.
Therapeutic T cell engineering
.
Nature.
2017
;
545
(
7655
):
423
-
431
.
4.
Cappell
KM
,
Sherry
RM
,
Yang
JC
, et al
.
Long-term follow-up of anti-CD19 chimeric antigen receptor T-cell therapy
.
J Clin Oncol.
2020
;
38
(
32
):
3805
-
3815
.
5.
Newick
K
,
O’Brien
S
,
Moon
E
,
Albelda
SM
.
CAR T cell therapy for solid tumors
.
Annu Rev Med.
2017
;
68
(
1
):
139
-
152
.
6.
Weber
EW
,
Maus
MV
,
Mackall
CL
.
The emerging landscape of immune cell therapies
.
Cell.
2020
;
181
(
1
):
46
-
62
.
7.
Tichelli
A
,
Beohou
E
,
Labopin
M
, et al;
Transplant Complications Working Party of the EBMT
.
Evaluation of second solid cancers after hematopoietic stem cell transplantation in European patients
.
JAMA Oncol.
2019
;
5
(
2
):
229
-
235
.
8.
Lee
DW
,
Santomasso
BD
,
Locke
FL
, et al
.
ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells
.
Biol Blood Marrow Transplant.
2019
;
25
(
4
):
625
-
638
.
9.
Brudno
JN
,
Kochenderfer
JN
.
Toxicities of chimeric antigen receptor T cells: recognition and management
.
Blood.
2016
;
127
(
26
):
3321
-
3330
.
10.
Rheingold
S
,
Neugut
A
,
Meadows
A
.
Secondary cancers: incidence, risk factors, and management
.
Cancer Med.
2003
;
6
:
2399
.
11.
Tsai
H-J
,
Wu
C-C
,
Tsai
C-R
,
Lin
S-F
,
Chen
L-T
,
Chang
JS
.
Second cancers in patients with neuroendocrine tumors
.
PLoS One.
2013
;
8
(
12
):
e86414
.
12.
Kohn
DB
,
Sadelain
M
,
Glorioso
JC
.
Occurrence of leukaemia following gene therapy of X-linked SCID
.
Nat Rev Cancer.
2003
;
3
(
7
):
477
-
488
.
13.
Hacein-Bey-Abina
S
,
Von Kalle
C
,
Schmidt
M
, et al
.
LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1
.
Science.
2003
;
302
(
5644
):
415
-
419
.
14.
Heslop
HE
,
Slobod
KS
,
Pule
MA
, et al
.
Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients
.
Blood.
2010
;
115
(
5
):
925
-
935
.
15.
Bollard
CM
,
Aguilar
L
,
Straathof
KC
, et al
.
Cytotoxic T lymphocyte therapy for Epstein-Barr virus+ Hodgkin’s disease
.
J Exp Med.
2004
;
200
(
12
):
1623
-
1633
.
16.
Zhou
X
,
Dotti
G
,
Krance
RA
, et al
.
Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation
.
Blood.
2015
;
125
(
26
):
4103
-
4113
.
17.
Zhou
X
,
Di Stasi
A
,
Tey
SK
, et al
.
Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene
.
Blood.
2014
;
123
(
25
):
3895
-
3905
.
18.
Bollard
CM
,
Tripic
T
,
Cruz
CR
, et al
.
Tumor-specific T-cells engineered to overcome tumor immune evasion induce clinical responses in patients with relapsed Hodgkin lymphoma
.
J Clin Oncol.
2018
;
36
(
11
):
1128
-
1139
.
19.
Louis
CU
,
Savoldo
B
,
Dotti
G
, et al
.
Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma
.
Blood.
2011
;
118
(
23
):
6050
-
6056
.
20.
Heczey
A
,
Louis
CU
,
Savoldo
B
, et al
.
CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma
.
Mol Ther.
2017
;
25
(
9
):
2214
-
2224
.
21.
Savoldo
B
,
Ramos
CA
,
Liu
E
, et al
.
CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients
.
J Clin Invest.
2011
;
121
(
5
):
1822
-
1826
.
22.
Ramos
CA
,
Rouce
R
,
Robertson
CS
, et al
.
In vivo fate and activity of second- versus third-generation CD19-specific CAR-T cells in B cell non-Hodgkin’s lymphomas
.
Mol Ther.
2018
;
26
(
12
):
2727
-
2737
.
23.
Ramos
CA
,
Ballard
B
,
Zhang
H
, et al
.
Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes
.
J Clin Invest.
2017
;
127
(
9
):
3462
-
3471
.
24.
Ramos
CA
,
Savoldo
B
,
Torrano
V
, et al
.
Clinical responses with T lymphocytes targeting malignancy-associated κ light chains
.
J Clin Invest.
2016
;
126
(
7
):
2588
-
2596
.
25.
Ramos
CA
,
Grover
NS
,
Beaven
AW
, et al
.
Anti-CD30 CAR-T cell therapy in relapsed and refractory Hodgkin lymphoma
.
J Clin Oncol.
2020
;
38
(
32
):
3794
-
3804
.
26.
Ahmed
N
,
Brawley
VS
,
Hegde
M
, et al
.
Human epidermal growth factor receptor 2 (HER2) -specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma
.
J Clin Oncol.
2015
;
33
(
15
):
1688
-
1696
.
27.
Ahmed
N
,
Brawley
V
,
Hegde
M
, et al
.
HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial
.
JAMA Oncol.
2017
;
3
(
8
):
1094
-
1101
.
28.
Bender
MA
,
Palmer
TD
,
Gelinas
RE
,
Miller
AD
.
Evidence that the packaging signal of Moloney murine leukemia virus extends into the gag region
.
J Virol.
1987
;
61
(
5
):
1639
-
1646
.
29.
Loew
R
,
Meyer
Y
,
Kuehlcke
K
, et al
.
A new PG13-based packaging cell line for stable production of clinical-grade self-inactivating γ-retroviral vectors using targeted integration
.
Gene Ther.
2010
;
17
(
2
):
272
-
280
.
30.
Mulligan
RC
.
Development of gene transfer technology
.
Hum Gene Ther.
2014
;
25
(
12
):
995
-
1002
.
31.
George
D
,
Mallery
P
.
Descriptive statistics. IBM SPSS statistics 23 step by step.
Routledge
;
2016
:
126
-
134
32.
Gray
RJ
.
A class of K-sample tests for comparing the cumulative incidence of a competing risk
.
Ann Stat.
1988
;
16
(
3
):
1141
-
1154
.
33.
Fine
JP
,
Gray
RJ
.
A proportional hazards model for the subdistribution of a competing risk
.
J Am Stat Assoc.
1999
;
94
(
446
):
496
-
509
.
34.
Bland
JM
,
Altman
DG
.
Survival probabilities (the Kaplan-Meier method)
.
BMJ.
1998
;
317
(
7172
):
1572
-
1580
.
35.
Vasen
HF
,
Ghorbanoghli
Z
,
Bourdeaut
F
, et al;
EU-Consortium Care for CMMR-D (C4CMMR-D)
.
Guidelines for surveillance of individuals with constitutional mismatch repair-deficiency proposed by the European Consortium “Care for CMMR-D” (C4CMMR-D)
.
J Med Genet.
2014
;
51
(
5
):
283
-
293
.
36.
Ripperger
T
,
Schlegelberger
B
.
Acute lymphoblastic leukemia and lymphoma in the context of constitutional mismatch repair deficiency syndrome
.
Eur J Med Genet.
2016
;
59
(
3
):
133
-
142
.
37.
Bear
AS
,
Morgan
RA
,
Cornetta
K
, et al
.
Replication-competent retroviruses in gene-modified T cells used in clinical trials: is it time to revise the testing requirements?
Mol Ther.
2012
;
20
(
2
):
246
-
249
.
38.
Cornetta
K
,
Duffy
L
,
Feldman
SA
, et al
.
Screening clinical cell products for replication competent retrovirus: the National Gene Vector Biorepository Experience
.
Mol Ther Methods Clin Dev.
2018
;
10
:
371
-
378
.
39.
Brown
MP
,
Topham
DJ
,
Sangster
MY
, et al
.
Thymic lymphoproliferative disease after successful correction of CD40 ligand deficiency by gene transfer in mice
.
Nat Med.
1998
;
4
(
11
):
1253
-
1260
.
40.
Shah
NN
,
Qin
H
,
Yates
B
, et al
.
Clonal expansion of CAR T cells harboring lentivector integration in the CBL gene following anti-CD22 CAR T-cell therapy
.
Blood Adv.
2019
;
3
(
15
):
2317
-
2322
.
41.
Fraietta
JA
,
Nobles
CL
,
Sammons
MA
, et al
.
Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells
.
Nature.
2018
;
558
(
7709
):
307
-
312
.
42.
Vargas
JE
,
Chicaybam
L
,
Stein
RT
,
Tanuri
A
,
Delgado-Cañedo
A
,
Bonamino
MH
.
Retroviral vectors and transposons for stable gene therapy: advances, current challenges and perspectives
.
J Transl Med.
2016
;
14
(
1
):
288
.
43.
Saha
S
,
Woodard
LE
,
Charron
EM
,
Welch
RC
,
Rooney
CM
,
Wilson
MH
.
Evaluating the potential for undesired genomic effects of the piggyBac transposon system in human cells
.
Nucleic Acids Res.
2015
;
43
(
3
):
1770
-
1782
.
44.
Bishop
DC
,
Clancy
LE
,
Simms
R
, et al
.
Development of CAR T-cell lymphoma in 2 of 10 patients effectively treated with piggyBac-modified CD19 CAR T cells
.
Blood.
2021
;
138
(
16
):
1504
-
1509
.
45.
Prommersberger
S
,
Reiser
M
,
Beckmann
J
, et al
.
CARAMBA: a first-in-human clinical trial with SLAMF7 CAR-T cells prepared by virus-free Sleeping Beauty gene transfer to treat multiple myeloma
.
Gene Ther.
2021
;
28
(
9
):
560
-
571
.
46.
Gregory
T
,
Cohen
AD
,
Costello
CL
, et al
.
Efficacy and safety of P-Bcma-101 CAR-T cells in patients with relapsed/refractory (r/r) multiple myeloma (MM)
.
Blood.
2018
;
132
(
suppl 1
):
1012
.
47.
Neelapu
SS
,
Nath
R
,
Munoz
J
, et al
.
ALPHA Study: ALLO-501 produced deep and durable responses in patients with relapsed/refractory non-Hodgkin’s lymphoma comparable to autologous CAR T
.
Blood.
2021
;
138
(
suppl 1
):
3878
.
48.
Sheridan
C
.
Off-the-shelf, gene-edited CAR-T cells forge ahead, despite safety scare
.
Nat Biotechnol.
2022
;
40
(
1
):
5
-
8
.
49.
Bonifant
CL
,
Jackson
HJ
,
Brentjens
RJ
,
Curran
KJ
.
Toxicity and management in CAR T-cell therapy
.
Mol Ther Oncolytics.
2016
;
3
:
16011
.
50.
Fischer
A
,
Hacein-Bey-Abina
S
.
Gene therapy for severe combined immunodeficiencies and beyond
.
J Exp Med.
2020
;
217
(
2
):
e20190607
.

Author notes

*

D.H.M.S. and I.N.M. contributed equally to this study.

Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.
Sign in via your Institution