T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer

Eric Tran, Paul F Robbins, Yong-Chen Lu, Todd D Prickett, Jared J Gartner, Li Jia, Anna Pasetto, Zhili Zheng, Satyajit Ray, Eric M Groh, Isaac R Kriley, Steven A Rosenberg, Eric Tran, Paul F Robbins, Yong-Chen Lu, Todd D Prickett, Jared J Gartner, Li Jia, Anna Pasetto, Zhili Zheng, Satyajit Ray, Eric M Groh, Isaac R Kriley, Steven A Rosenberg

Abstract

We identified a polyclonal CD8+ T-cell response against mutant KRAS G12D in tumor-infiltrating lymphocytes obtained from a patient with metastatic colorectal cancer. We observed objective regression of all seven lung metastases after the infusion of approximately 1.11×1011 HLA-C*08:02-restricted tumor-infiltrating lymphocytes that were composed of four different T-cell clonotypes that specifically targeted KRAS G12D. However, one of these lesions had progressed on evaluation 9 months after therapy. The lesion was resected and found to have lost the chromosome 6 haplotype encoding the HLA-C*08:02 class I major histocompatibility complex (MHC) molecule. The loss of expression of this molecule provided a direct mechanism of tumor immune evasion. Thus, the infusion of CD8+ cells targeting mutant KRAS mediated effective antitumor immunotherapy against a cancer that expressed mutant KRAS G12D and HLA-C*08:02.

Figures

Figure 1. Adoptive Transfer of KRAS G12D–Specific…
Figure 1. Adoptive Transfer of KRAS G12D–Specific T Cells
Panel A shows the flow cytometric analysis of the effector function of tumor-infiltrating lymphocytes in the infusion product with the use of intracellular cytokine staining (including interferon-γ [IFN-γ], tumor necrosis factor [TNF], and interleukin-2 [IL-2]), and cell-surface mobilization of the degranulation marker CD107a after 6-hour coculture with autologous dendritic cells incubated overnight with wild-type (WT) KRAS or KRAS G12D peptides consisting of 24 amino acids. Data are gated on CD8+ T cells. Pie charts represent the percentages of CD8+ cells that expressed the indicated number of effector functions. Panel B shows contrast-enhanced computed tomographic scans of the chest of Patient 4095 before and approximately 6 weeks and 9 months after the infusion of 1.48×1011 tumor-infiltrating lymphocytes; at least 75% of these cells were specific for mutant KRAS G12D. Arrows highlight lung lesions before and after therapy. Shown are four of seven lesions; the remaining three lesions (not shown) had completely regressed at 9 months.
Figure 2. In Vivo Persistence and Reactivity…
Figure 2. In Vivo Persistence and Reactivity Profile of KRAS G12D–Specific T-Cell Clones in the Infusion Product
Panel A shows the results of deep sequencing of the variable region of the T-cell receptor (TCR) beta chain to measure the frequency of each of the four identified KRAS G12D–reactive T-cell clones in the infusion product (Rx1), in three metastatic lung samples before cell transfer (designated Tu-1, Tu-2A, and Tu-2B), in the one progressing lesion after cell therapy (designated Tu-Pro), and in the peripheral blood of Patient 4095 before and at various times after cell therapy. T-cell receptors are identified according to their gene names (TRBV5-6 and TRBV10-02, with A, B, and C denoting different T-cell receptor clonotypes that share the same T-cell receptor beta variable gene family). The numbers in parentheses indicate the rank of the T-cell receptor sequence in the given sample. A circle with an X and ND denote not detected (

Figure 3. Analysis of the Frequency of…

Figure 3. Analysis of the Frequency of HLA-C Alleles in Tumor Samples

Shown is the…

Figure 3. Analysis of the Frequency of HLA-C Alleles in Tumor Samples
Shown is the frequency of the two HLA-C alleles in a representative metastatic lesion resected from Patient 4095 before cell therapy (Tu-1) and in the progressing lesion (Tu-Pro). The Tu-1 and Tu-Pro samples were estimated to contain 53% and 34% tumor, respectively, and thus contained some normal cells that also contributed to the calculation of the HLA-C allele frequency. As shown, the progressing lesion harbored cells with a genetic loss of the HLA-C*08:02 allele, which provided a direct mechanism of immune evasion by the tumor, since the infused KRAS G12D–reactive T cells require this molecule for direct tumor recognition. The I bars represent standard errors. The P value for the comparison of the allele frequency of a metastatic lesion before therapy and the progressing lesion after therapy was calculated with the use of the Wilcoxon matched-pairs signed-rank test.
Figure 3. Analysis of the Frequency of…
Figure 3. Analysis of the Frequency of HLA-C Alleles in Tumor Samples
Shown is the frequency of the two HLA-C alleles in a representative metastatic lesion resected from Patient 4095 before cell therapy (Tu-1) and in the progressing lesion (Tu-Pro). The Tu-1 and Tu-Pro samples were estimated to contain 53% and 34% tumor, respectively, and thus contained some normal cells that also contributed to the calculation of the HLA-C allele frequency. As shown, the progressing lesion harbored cells with a genetic loss of the HLA-C*08:02 allele, which provided a direct mechanism of immune evasion by the tumor, since the infused KRAS G12D–reactive T cells require this molecule for direct tumor recognition. The I bars represent standard errors. The P value for the comparison of the allele frequency of a metastatic lesion before therapy and the progressing lesion after therapy was calculated with the use of the Wilcoxon matched-pairs signed-rank test.

Source: PubMed

3
Subskrybuj