Weekly Digests
‹ Back to December

Intentional mutagenesis: making cancer worse to make it better

December 20, 2017

Although it seems counterintuitive that shutting down DNA repair mechanisms in cancer could actually improve prognosis, that’s exactly the concept that Germano et al. present in a letter published in Nature. They propose that when mismatch repair (MMR) mechanisms are disrupted in colorectal tumors, new mutations, and thus new potential neoantigens, continually arise, renewing the tumors’ susceptibility to attack by the host immune system.

In the clinic, colorectal cancers (CRCs) are often early onset and progress rapidly, yet they have a peculiarly favorable prognosis and respond well to immune checkpoint blockade. Previously, researchers hypothesized that the high mutational burden in these cancer might lead to an increased number of neoantigens, making such tumors vulnerable to attack by the immune system. Across multiple settings, however, mutation burden alone has not been a reliable predictor of prognosis. To study whether MMR mechanisms actively contributed to the clinical features of CRCs, Germano et al. turned their focus to the MLH1 protein, an essential protein in MMR complexes.

Using the CRISPR/Cas9 system, the researchers inactivated Mlh1 in CT26 and MC38 cancer cell lines, which caused MMR deficiency. The MMR-deficient cancer cell lines proliferated well in vitro and when injected into immunocompromised mice. However, when injected (either subcutaneously or orthotopically) into immunocompetent syngeneic mice, they failed to grow or grew very poorly in locations where the MMR-proficient cancer cell lines grew well, indicating that the immune system was involved in tumor suppression. Treatment with anti-CD8 antibody abrogated tumor control, indicating that CD8+ T cells were instrumental in the effect.

To mimic a clinical setting of established tumors being treated with checkpoint inhibitors, the researchers grew MMR-deficient or -proficient tumors in immunocompromised mice and transplanted fragments into immunocompetent syngeneic mice. When the transplanted tumors reached target size, some were treated with a combination of anti-PD-1 and anti-CTLA-4. While MMR-proficient tumors grew well and were insensitive to checkpoint therapy, MMR-deficient tumors grew more slowly, responded to checkpoint therapy, and had higher CD8+ T cell infiltration.

RNA sequencing of tumors revealed that while MMR-proficient tumors showed a stable number of mutations over time, MMR-deficient tumors had an increasing number of mutations, indicating that new mutations, and thus new predicted neoantigens, were continually emerging over time. In theory, the emergence of new neoantigens would allow the immune system to spot and attack new targets as they arise, rather than becoming ineffective once immunoediting has occurred.

In an attempt to exploit MMR-deficiency as a possible immunotherapeutic strategy, Germano et al. turned to temozolomide (TMZ), an FDA approved chemotherapeutic agent known to trigger DNA damage that can result in inactivation of mismatch repair. Using MC38 tumor cells, the team successfully generated TMZ-resistant cells carrying a de novo Mlh1 deletion which was not present in the MMR-proficient parental cells before TMZ treatment. This indicated that mismatch repair had successfully been inactivated; confirming this result, the altered MC38 cells were unable to form tumors in immunocompetent mice.

To determine whether this effect of TMZ would be relevant in humans, the researchers analyzed a collection of colorectal cancer cell lines and generated TMZ-resistant lines from the subset that were initially sensitive to TMZ. In line with data from mouse models, human CRC cells with altered MMR mechanisms showed high levels of neoantigens, which increased over time. The researchers were also able to obtain tissue samples from five patients who had achieved partial responses or prolonged stabilization following TMZ-based chemotherapy. These samples revealed that after treatment, three of the five patients’ cancers upregulated MGMT, the main enzyme responsible for repairing TMZ-damaged DNA. The other two maintained low MGMT levels and were found to have a high mutational burden and mutations in an MMR-related gene that were not present before TMZ therapy.

Germano et al. conclude that silencing mismatch repair genes dynamically increases the number of de novo mutations and neoantigens in cancer cells, which can result in continually expanding and enhanced immune responses over time.

by Lauren Hitchings

References:

Germano G., Lamba S., Rospo G., Barault L., Magrì A., Maione F., Russo M., Crisafulli G., Bartolini A., Lerda G., Siravegna G., Mussolin B., Frapolli R., Montone M., Morano F., de Braud F., Amirouchene-Angelozzi N., Marsoni S., D'Incalci M., Orlandi A., Giraudo E., Sartore-Bianchi A., Siena S., Pietrantonio F., Di Nicolantonio F., Bardelli A. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature. 2017 Dec 7.

In the Spotlight...

Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma

In a Phase II clinical trial, 101 patients with relapsed/refractory large B-cell lymphoma were treated with axicabtagene ciloleucel, an autologous CD19 CAR (CD28) T cell therapy, following lymphodepletion with fludarabine and cyclophosphamide. The objective and complete response rates were 82% and 54% at 6 months, and the overall survival rate was 52% at 18 months. Severe toxicities included myelosuppression, cytokine release syndrome, and neurological events. Response was associated with higher CAR T cell levels in the blood.

Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas

In a Phase IIa clinical trial, 28 patients with relapsed/refractory diffuse large B-cell lymphoma or follicular lymphoma were treated with autologous CD19 CAR (41BB) T cells following investigator-chosen lymphodepletion. Overall response rate was 64%, and complete remission rate was 57%. Remissions were durable. Severe toxicities included cytokine release syndrome and encephalopathy, which fully resolved in all but one case. Some patients experienced B-cell and immunoglobulin recovery, despite having detectable levels of CAR T cells in the blood.

Inhibition of AKT signaling uncouples T cell differentiation from expansion for receptor-engineered adoptive immunotherapy

To prevent undesirable differentiation during production of CAR- or TCR-engineered human T cells, Klebanoff et al. found that the addition of an AKT inhibitor preserved minimally differentiated CD62L-expressing populations and promoted favorable transcriptional and metabolic properties while being compatible with efficient viral transduction and expansion. CAR T cells prepared with AKT inhibitors improved survival in mice compared to conventionally engineered cells, prompting integration of this strategy into a clinical trial.

Human CD26(high) T cells elicit tumor immunity against multiple malignancies via enhanced migration and persistence

Bailey et al. found that differential expression of CD26, an enzyme that plays a role in T cell costimulation and binding to extracellular matrix proteins, defines three human CD4+ T cell subsets: regulatory (CD26neg), naive (CD26int), and stem memory (CD26high). When used in adoptive cell transfer, CD26high T cells exhibit persistence and promote regression of multiple tumors due to enhanced cytotoxicity, high polyfunctionality, expansive chemokine receptor repertoire, increased co-inhibitory and co-stimulatory markers, and resistance to apoptosis.

Human PBMC-transferred murine MHC class I/II-deficient NOG mice enable long-term evaluation of human immune responses

Yaguchi et al. created NOG mice that are MHC class I- and class II-deficient (NOG-dKO) to minimize the effects of graft-versus-host-disease, allowing long-term survival and extended observation of human immune responses following PBMC transplantation. The model could be used to study antigen-specific human B and T cell responses, as well as adoptive cell immunotherapies. However, antigen-presenting cells have short longevity and many immune cell types (including macrophages, Tregs, MDSCs, and NK cells) are not well supported.

A simple, clinically relevant therapeutic vaccine shows long-term protection in an aggressive, delayed-treatment B lymphoma model

Pradhan et al. demonstrated that a vaccine strategy using a lysate of the diffuse large B-cell lymphoma A20 cell line adjuvanted with the NKT cell agonist α-GalCer was effective and durable against pre-established B ALL lymphoma in mice. Efficacy was dependent on CD8+ T cells and was associated with increases in NKT and NK cells and decreased granulocytic MDSCs and Tregs. Enhanced germinal center reaction and magnitude of class-switching to a Th1 type humoral response were identified as biomarkers of durable response.

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.