Weekly Digests
‹ Back to July

To maximize immune response, leave the lymph node alone

July 11, 2018

In clinical practice, radiation oncologists frequently perform elective nodal irradiation (ENI) to irradiate clinically uninvolved tumor-draining lymph nodes (TDLN) in addition to the localized tumor in order to preemptively treat potential nodal micrometastases. However, it is unclear how the irradiation of TDLN affects adaptive immune response and the efficacy of radiation therapy in combination with immune checkpoint blockade (ICB). Marciscano et al., in their recent paper published in Clinical Cancer Research, attempted to answer this question by developing a preclinical model of ENI and comparing the immunological effects of tumor-only stereotactic radiotherapy alone (RT) and tumor radiotherapy + ENI (RT + ENI), as monotherapies or in combination with ICB.

To develop their preclinical model, the team utilized cone-beam computed tomography imaging using the small animal radiation research platform (SARRP) on C57BL/6J mice with implanted B16 melanoma or MC38 colorectal carcinoma to perform image-guided stereotactic radiotherapy on the tumor or the tumor and the TDLN. The absence of micrometastatic disease in the TDLN at the time of radiotherapy was confirmed by a pathologist.

Marciscano et al. irradiated mice 11 days after tumor implantation and found that RT significantly increased the tumor infiltration of CD8+ effector T cells (Teffs), Tregs, and immunosuppressive CD11b+Gr-1hi myeloid cells, while RT + ENI led to only a slight increase in Teffs and did not significantly affect the other two cell subsets. Both treatments increased the CD8+ Teff/Treg ratio relative to non-irradiated controls, although the effect was significantly greater with RT than with RT + ENI. The increased intratumoral infiltration of Teffs following RT compared with RT + ENI was also confirmed via adoptive transfer of OVA-specific CD8+ T cells (OT-1) into mice with OVA-expressing tumors.

Digging deeper into the mechanism behind the differential immunological effects of the two radiotherapy treatments, Marciscano et al. observed that RT led to upregulation of intratumoral CXCR3- and CCR5-associated chemokines (CXCL10, CCL3, CCL5, etc.), which play a role in recruitment of CD8+ Teffs into the tumor microenvironment (TME). Chemokine upregulation was lower after RT + ENI, which may explain the lower Teff accumulation in the TME following this treatment. It is worth noting that RT + ENI did not negatively affect T cell priming within the TDLN. In fact, adoptive transfer experiments demonstrated that RT + ENI resulted in significantly higher proportion of cytokine-producing OT-1 cells in the TDLN compared with RT. However, this increase in TDLN Teffs did not translate to increased migration into the tumor due to decreased chemokine expression.

Next, the team sought to examine whether RT and RT + ENI differentially impact survival when used as monotherapy or combined with ICB. As monotherapies, both RT and RT + ENI prolonged survival (41 and 25 days, respectively) compared with control, but neither therapy led to complete tumor regression or long-term survival. Combining either radiotherapy strategy with anti-PD-1 improved survival compared with anti-PD-1 alone (~45 days versus 25 days) and led to a complete response rate of approximately 30% for both RT/anti-PD-1 and RT + ENI/anti-PD-1. Anti-CTLA-4 monotherapy, which partially depletes Tregs, resulted in 29% complete tumor regression rate and median survival of 44 days. Combination RT + ENI/anti-CTLA-4 further improved the outcome, with survival of 62 days and complete response in 43% of mice. However, the best observed outcome was with RT/anti-CTLA-4 treatment, which resulted in complete response rate of 86% and median survival rate that had not been reached by day 90.

Using immunophenotyping to quantify the differences in immunological effects of the different treatment groups, the researchers found that the absolute number of CD8+ Teffs and the Teff/Treg ratio in the tumor positively correlated with survival. Not surprisingly, the RT/anti-CTLA-4 treatment group demonstrated the highest Teff/Treg ratio and CD8+ Teff infiltration in the tumor. Interestingly, while anti-CTLA-4 decreased intratumoral Tregs when used as monotherapy or in combination with RT, combining anti-CTLA-4 with RT + ENI increased intratumoral Tregs, negatively impacting survival.

Surviving mice (found only in combination treatment groups) were rechallenged six months after initial tumor implantation to examine the formation of memory response. Mice treated with anti-CTLA-4 in combination with either radiation strategy had a more robust memory response than mice treated with anti-PD-1 in combination with either radiation treatment (100% versus 50%, respectively). Stratifying memory response by radiation type (combined with either ICB) revealed no significant difference in survival. Together, these results indicate that while the choice of radiotherapy treatment impacts initial response and survival, long-term protection is more dependent on the choice of ICB.

Overall, Marciscano et al. demonstrate that irradiation of the clinically uninvolved TDLN attenuates the immunomodulatory benefit of stereotactic irradiation of the tumor, negatively impacting adaptive immune response via reduced chemokine expression and reduced trafficking of CD8+ Teffs into the tumor. Moreover, synergy of tumor irradiation with anti-CTLA-4 is abrogated. The preclinical data presented in this study suggests that irradiation of TDLN should be carefully reconsidered in order to improve the efficacy of radiotherapy/immunotherapy combination treatment in the clinic.

by Anna Scherer

References:

Marciscano A.E., Ghasemzadeh A., Nirschl T.R., Theodros D., Kochel C.M., Francica B.J., Muroyama Y., Anders R.A., Sharabi A.B., Velarde E., Mao W., Chaudhary K.R., Chaimowitz M.G., Wong J., Selby M., Thudium K.B., Korman A.J., Ulmert D., Thorek D.L., DeWeese T.L., Drake C.G. Elective nodal irradiation attenuates the combinatorial efficacy of stereotactic radiation therapy and immunotherapy. Clin Cancer Res. 2018 Jun 13.

In the Spotlight...

Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity

Following the observation that TIGIT, a known checkpoint receptor on T cells, was upregulated on intratumoral NK cells in colon cancer patients, Zhang et al. found that TIGIT upregulation in mice was associated with NK cell exhaustion and tumor progression. Genetic TIGIT deficiency or antibody blockade alleviated NK cell exhaustion, slowed tumor growth independent of adaptive immunity, and generated long-term memory. Interestingly, TIGIT blockade enhanced T cell immunity in an NK cell-dependent manner, and mono- or combination therapies with anti-TIGIT and/or anti-PD-L1 were dependent on the presence of NK cells.

Induction and transcriptional regulation of the co-inhibitory gene module in T cells

Using single-cell expression profiling of CD4+ and CD8+ tumor-infiltrating T cells, Chihara and Madi et al. identified a co-inhibitory gene module that includes the well-known receptors PD-1, TIM-3, LAG-3, and TIGIT, as well as two novel surface molecules: activated protein C receptor (PROCR) and podoplanin (PDPN). IL-27 was found to upregulate transcription factors PRDM1 and c-MAF, which in turn redundantly upregulated the co-inhibitory module. Double knockout of PRDM1 and c-MAF almost eliminated the expression of co-inhibitory receptors, increased the presence of non-exhausted effector T cells, and reduced tumor growth in mice.

Critical role of integrin CD11c in splenic dendritic cell capture of missing-self CD47 cells to induce adaptive immunity

Wu et al. found that CD11c (integrin αX), the defining marker for dendritic cells (DCs), and its partner, integrin β2 (Itgb2), were critical for splenic cDC2 capture of cells deficient in self-marker CD47 and for DC activation in a manner dependent on integrin-signaling regulator Talin1. In vivo, Talin1-deficient cDC2s were ineffective in inducing TFH and T cell-dependent B cell responses to cells lacking CD47. CD11b (integrin αM), which is also expressed on cDC2s, was not required for capture of CD47-deficient cells, but partially compensated for the absence of CD11c. The activating receptor for the CD11c/Itgb2 complex is unknown.

Development of MGD007, a gpA33 x CD3 bispecific DART(R) protein for T-cell immunotherapy of metastatic colorectal cancer

Moore et al. identified gpA33 as a target found on colorectal cancer stem cell lines as well as on primary and metastatic colorectal tumor specimens, and describe preclinical analyses supporting the clinical development of a gpA33 T cell re-directing antibody. The dual-affinity retargeting (DART) molecule used a three-chain platform design, including a half-life-extending Fc region, and demonstrated efficient colorectal cancer cell lysis in vitro and in vivo. Multiple T cell subsets (CD8+, CD4+, and CD4+FOXP3+ Treg) were active in cytolysis. Prolonged antigen exposure drove expansion and loss of cytokine production, but cytolytic activity was retained.

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.