Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy. / Chamberlain, Christopher Aled; Bennett, Eric Paul; Kverneland, Anders Handrup; Svane, Inge Marie; Donia, Marco; Met, Özcan.

In: Molecular Therapy - Oncolytics, Vol. 24, 2022, p. 417-428.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Chamberlain, CA, Bennett, EP, Kverneland, AH, Svane, IM, Donia, M & Met, Ö 2022, 'Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy', Molecular Therapy - Oncolytics, vol. 24, pp. 417-428. https://doi.org/10.1016/j.omto.2022.01.004

APA

Chamberlain, C. A., Bennett, E. P., Kverneland, A. H., Svane, I. M., Donia, M., & Met, Ö. (2022). Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy. Molecular Therapy - Oncolytics, 24, 417-428. https://doi.org/10.1016/j.omto.2022.01.004

Vancouver

Chamberlain CA, Bennett EP, Kverneland AH, Svane IM, Donia M, Met Ö. Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy. Molecular Therapy - Oncolytics. 2022;24:417-428. https://doi.org/10.1016/j.omto.2022.01.004

Author

Chamberlain, Christopher Aled ; Bennett, Eric Paul ; Kverneland, Anders Handrup ; Svane, Inge Marie ; Donia, Marco ; Met, Özcan. / Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy. In: Molecular Therapy - Oncolytics. 2022 ; Vol. 24. pp. 417-428.

Bibtex

@article{57799c4645534c81b87497d72e2ad3b3,
title = "Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy",
abstract = "Adoptive T cell therapy (ACT) with expanded tumor-infiltrating lymphocytes (TIL) can induce durable responses in cancer patients from multiple histologies, with response rates of up to 50%. Antibodies blocking the engagement of the inhibitory receptor programmed cell death protein 1 (PD-1) have been successful across a variety of cancer diagnoses. We hypothesized that these approaches could be combined by using CRISPR-Cas9 gene editing to knock out PD-1 in TILs from metastatic melanoma and head-and-neck, thyroid, and colorectal cancer. Non-viral, non-plasmid-based PD-1 knockout was carried out immediately prior to the traditional 14-day TIL-based ACT rapid-expansion protocol. A median 87.53% reduction in cell surface PD-1 expression was observed post-expansion and confirmed at the genomic level. No off-target editing was detected, and PD-1 knockout had no effect on final fold expansion. Edited cells exhibited few phenotypic differences and matched control functionality. Pre-clinical-scale results were confirmed at a clinical scale by generating a PD-1-deficient TIL product using the good manufacturing practice facilities, equipment, procedures, and starting material used for standard patient treatment. Our results demonstrate that simple, non-viral, non-plasmid-based CRISPR-Cas9 methods can be feasibly adopted into a TIL-based ACT protocol to produce treatment products deficient in molecules such as PD-1, without any evident negative effects.",
keywords = "adoptive cell therapy, CRISPR-Cas9, gene editing, immunotherapy, PD-1, TIL therapy, tumor-infiltrating lymphocytes",
author = "Chamberlain, {Christopher Aled} and Bennett, {Eric Paul} and Kverneland, {Anders Handrup} and Svane, {Inge Marie} and Marco Donia and {\"O}zcan Met",
note = "Publisher Copyright: {\textcopyright} 2022 The Authors",
year = "2022",
doi = "10.1016/j.omto.2022.01.004",
language = "English",
volume = "24",
pages = "417--428",
journal = "Molecular Therapy - Oncolytics",
issn = "2372-7705",
publisher = "Nature Publishing Group",

}

RIS

TY - JOUR

T1 - Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy

AU - Chamberlain, Christopher Aled

AU - Bennett, Eric Paul

AU - Kverneland, Anders Handrup

AU - Svane, Inge Marie

AU - Donia, Marco

AU - Met, Özcan

N1 - Publisher Copyright: © 2022 The Authors

PY - 2022

Y1 - 2022

N2 - Adoptive T cell therapy (ACT) with expanded tumor-infiltrating lymphocytes (TIL) can induce durable responses in cancer patients from multiple histologies, with response rates of up to 50%. Antibodies blocking the engagement of the inhibitory receptor programmed cell death protein 1 (PD-1) have been successful across a variety of cancer diagnoses. We hypothesized that these approaches could be combined by using CRISPR-Cas9 gene editing to knock out PD-1 in TILs from metastatic melanoma and head-and-neck, thyroid, and colorectal cancer. Non-viral, non-plasmid-based PD-1 knockout was carried out immediately prior to the traditional 14-day TIL-based ACT rapid-expansion protocol. A median 87.53% reduction in cell surface PD-1 expression was observed post-expansion and confirmed at the genomic level. No off-target editing was detected, and PD-1 knockout had no effect on final fold expansion. Edited cells exhibited few phenotypic differences and matched control functionality. Pre-clinical-scale results were confirmed at a clinical scale by generating a PD-1-deficient TIL product using the good manufacturing practice facilities, equipment, procedures, and starting material used for standard patient treatment. Our results demonstrate that simple, non-viral, non-plasmid-based CRISPR-Cas9 methods can be feasibly adopted into a TIL-based ACT protocol to produce treatment products deficient in molecules such as PD-1, without any evident negative effects.

AB - Adoptive T cell therapy (ACT) with expanded tumor-infiltrating lymphocytes (TIL) can induce durable responses in cancer patients from multiple histologies, with response rates of up to 50%. Antibodies blocking the engagement of the inhibitory receptor programmed cell death protein 1 (PD-1) have been successful across a variety of cancer diagnoses. We hypothesized that these approaches could be combined by using CRISPR-Cas9 gene editing to knock out PD-1 in TILs from metastatic melanoma and head-and-neck, thyroid, and colorectal cancer. Non-viral, non-plasmid-based PD-1 knockout was carried out immediately prior to the traditional 14-day TIL-based ACT rapid-expansion protocol. A median 87.53% reduction in cell surface PD-1 expression was observed post-expansion and confirmed at the genomic level. No off-target editing was detected, and PD-1 knockout had no effect on final fold expansion. Edited cells exhibited few phenotypic differences and matched control functionality. Pre-clinical-scale results were confirmed at a clinical scale by generating a PD-1-deficient TIL product using the good manufacturing practice facilities, equipment, procedures, and starting material used for standard patient treatment. Our results demonstrate that simple, non-viral, non-plasmid-based CRISPR-Cas9 methods can be feasibly adopted into a TIL-based ACT protocol to produce treatment products deficient in molecules such as PD-1, without any evident negative effects.

KW - adoptive cell therapy

KW - CRISPR-Cas9

KW - gene editing

KW - immunotherapy

KW - PD-1

KW - TIL therapy

KW - tumor-infiltrating lymphocytes

U2 - 10.1016/j.omto.2022.01.004

DO - 10.1016/j.omto.2022.01.004

M3 - Journal article

C2 - 35141398

AN - SCOPUS:85123602167

VL - 24

SP - 417

EP - 428

JO - Molecular Therapy - Oncolytics

JF - Molecular Therapy - Oncolytics

SN - 2372-7705

ER -

ID: 291603968