Abstract
Immunotherapy failures can result from the highly suppressive tumour microenvironment that characterizes aggressive forms of cancer such as recurrent glioblastoma (rGBM)1,2. Here we report the results of a first-in-human phase I trial in 41 patients with rGBM who were injected with CAN-3110—an oncolytic herpes virus (oHSV)3. In contrast to other clinical oHSVs, CAN-3110 retains the viral neurovirulence ICP34.5 gene transcribed by a nestin promoter; nestin is overexpressed in GBM and other invasive tumours, but not in the adult brain or healthy differentiated tissue4. These modifications confer CAN-3110 with preferential tumour replication. No dose-limiting toxicities were encountered. Positive HSV1 serology was significantly associated with both improved survival and clearance of CAN-3110 from injected tumours. Survival after treatment, particularly in individuals seropositive for HSV1, was significantly associated with (1) changes in tumour/PBMC T cell counts and clonal diversity, (2) peripheral expansion/contraction of specific T cell clonotypes; and (3) tumour transcriptomic signatures of immune activation. These results provide human validation that intralesional oHSV treatment enhances anticancer immune responses even in immunosuppressive tumour microenvironments, particularly in individuals with cognate serology to the injected virus. This provides a biological rationale for use of this oncolytic modality in cancers that are otherwise unresponsive to immunotherapy (ClinicalTrials.gov: NCT03152318).
Subject terms: Phase I trials, CNS cancer
Treatment with the oncolytic herpes virus CAN-3110 is associated with improved survival responses in patients with recurrent glioblastoma, particularly in individuals who are seropositive for HSV1.
Main
High-grade gliomas (HGGs) are central nervous system tumours of glial origin with highly malignant morphologic and genetic features5,6. Among these, GBM is characterized by the worst outcome in terms of survival, with rapid recurrence after neurosurgical resection and chemoradiation7. Recurrent HGG (rHGG), including recurrent GBM (rGBM), is characterized by rapid neurological morbidity and survival of less than 10 months8. Although much is known of the genetics, cellular composition and evolution of HGG/GBM, this has not translated into successful therapies. Traditional immunotherapy has also been ineffective in rHGG/rGBM1. This is thought to be due to the scarcity of infiltrating antitumour lymphocytes caused by a highly immunosuppressive tumour microenvironment (TME), defining these tumours as ‘lymphocyte depleted’2. For rGBMs and several other highly immunosuppressive solid cancers, there is a need to find treatment modalities that can convert the TME into one that is more amenable to immunotherapy and immune activation.
Oncolytic viruses are a form of immunotherapy in which oncolytic-virus-induced oncolysis alters the TME, promoting proinflammatory pathways, activating resident and newly recruited immune cells through exposure of viral and possibly tumour antigens9–13. Several oncolytic viruses have been and continue to be tested in oncology, with one approved as a single-agent intralesional injection into melanoma14 and a second one approved for injection into rGBM in Japan15–17. Notably, several early-phase oncolytic-virus clinical trials for HGG have been published in recent high-profile literature17–23. Yet, immunological profiling of rGBMs treated with oncolytic viruses in numbers sufficient to correlate with a therapeutic outcome has been lacking.
Here we report safety data for a first-in-human phase I clinical trial in 41 patients with rHGG/rGBM who were treated with CAN-3110—an oncolytic virus derived from herpes simplex virus type 1 (oncolytic HSV (oHSV); ClinicalTrials.gov: NCT03152318). We found that patients whose survival response after CAN-3110 was the longest were characterized by positive HSV1 serology with CAN-3110 clearance from infected tumour, differences in T cell clonotype metrics, and tumour transcriptomic signatures associated with immune activation programs. These findings provide human immunological and biological evidence supporting intralesional oncolytic treatment modalities to change the immunosuppressive TME into one that is more favourable for immunotherapy, providing broad relevance for the therapy of many solid cancers that are otherwise impervious to immune rejection.
Safety of CAN-3110 in patients with rHGG/rGBM
Most clinical oHSVs to date have deleted or removed the viral gene encoding ICP34.5 (refs. 3,4); although ICP34.5 enables robust replication of HSV in infected cells24,25, it is also responsible for neurotoxicity in mice26. To take advantage of ICP34.5’s functions that enhance viral replication/persistence and minimize neurotoxicity, CAN-3110 (former designation, rQNestin34.5v.2) was engineered to express a copy of the viral ICP34.5 gene under transcriptional control of the promoter for nestin, restricting viral replication and virulence to HGG/GBM cells3,4. To further ensure safety for initial use in humans, a multi-cohort clinical trial design was implemented (Extended Data Fig. 1a). Moreover, to ensure that the injections occurred in tumour, intraoperative MRI guidance was used to visualize the injections (Extended Data Fig. 1b,c and Supplementary Methods). A total of 41 patients with rHGG/rGBM (42 interventions, see the note on participant 042/054 in the Supplementary Methods; Extended Data Tables 1 and 2) were recruited to the trial. The patients were enrolled at their first (n = 18), second (n = 9) or third (n = 3) recurrence for cohorts 1–9 and at the first (n = 5), second (n = 3), third (n = 1) or fourth (n = 3) recurrence for cohort 10 (Extended Data Table 3). Tumour genomic data were typical for a rHGG/rGBM population (Extended Data Fig. 2), including the presence of mutations in the CDKN2A/B (encoding p16) tumour suppressor pathway, previously shown to complement viral replication of oHSVs, such as CAN-3110, with defects in the viral ribonucleotide reductase function27. Serious adverse events, consisting of seizures requiring hospitalization and intervention, were observed in two patients, but there were no dose-limiting toxicities or clinical/pathological evidence of ICP34.5-induced HSV1 encephalitis/meningitis (Tables 1 and 2 and Extended Data Table 4). Thus, these data indicate the relative human safety of CAN-3110 at all tested doses despite the presence of the HSV1 ICP34.5 neurovirulence gene.
Extended Data Table 1.
(related to Sub-heading, Safety of CAN-3110 in rHGG/rGBM patients)
*One subject in cohort 9 (subject 042) was re-treated as part of cohort 10 (subject 054). See explanation in Supplemental Text.
Extended Data Table 2.
(related to Sub-heading, Safety of CAN-3110 in rHGG/rGBM patients)
*One subject in cohort 9 (subject 042) was re-treated as part of cohort 10 (subject 054). See explanation in Supplemental Text.
Extended Data Table 3.
(related to Sub-headings, Safety of CAN-3110 in rHGG/rGBM patients and to Sub-Heading, HSV1 serology predicts efficacy)
*One subject in cohort 9 (subject 042) was re-treated as part of cohort 10 (subject 054). See explanation in Supplemental Text. †Multifocal staining pattern could be caused by tissue fragmentation.
Table 1.
Category | CTC grade 1 | CTC grade 2 |
---|---|---|
Blood and lymphatic systems disorders | ||
Low eosinophil count | 1 | 0 |
General disorders and administration site conditions | ||
Fatigue | 1 | 0 |
Fever | 3 | 0 |
Investigations | ||
Alanine aminotransferase increased | 1 | 0 |
Lymphocyte count decreased | 1 | 0 |
Platelet count decreased | 1 | 0 |
Musculoskeletal and connective tissue disorders | ||
Muscle weakness—lower limb | 1 | 0 |
Muscle weakness—upper limb | 1 | 0 |
Nervous systems disorders | ||
Cerebral oedema | 2 | 1 |
Headache | 0 | 1 |
Expressive aphasia | 1 | 0 |
Left leg numbness | 0 | 1 |
Left visual field defect | 0 | 1 |
Right arm joint position sense loss | 1 | 0 |
Seizure | 0 | 1 |
Speech | 0 | 1 |
Events reported as of 18 April 2022.
Table 2.
Case | Dose cohort | Days after CAN-3110 | Category | Adverse event | CTC grade | Relation to CAN-3110 | SUSAR |
---|---|---|---|---|---|---|---|
033 |
Arm A 3 × 109 |
16 | Nervous system disorders | Seizure | 3 | Possible | N |
033 |
Arm A 3 × 109 |
21 | Nervous system disorders | Cerebral haematoma | 3 | Possible | N |
046 |
Arm A 1 × 109 (2 ml) |
2 | Nervous system disorders | Seizure | 3 | Possible | N |
046 |
Arm A 1 × 109 (2 ml) |
3 | Nervous system disorders | Muscle weakness, left-sided | 3 | Possible | N |
Nervous system disorders | Muscle weakness, facial muscle | 3 | Possible | N |
SUSAR, suspected unexpected serious adverse reaction.
Extended Data Table 4.
(related to Sub-heading, Safety of CAN-3110 in rHGG/rGBM patients).
All grade 3 s are reported in Table 1.
HSV1 serology predicts efficacy
We tried to determine whether there were patients who benefited the most from treatment. Notably, 9 out of 41 patients (22%) had tumours associated with reduced survival28–30, such as depth (insular, thalamic), multifocality/multicentricity or bilateral laterality. In these latter cases, only one of the tumours or one hemispheric side of tumour was injected. Notably, patients like these are not routinely eligible for clinical trials, compounding the difficulty in comparing to historical clinical trial data. The estimated median overall survival (mOS) of the entire rHGG/rGBM group was 11.6 months (95% confidence interval (CI) = 7.8–14.9 months) (Fig. 1a). On the basis of the latest WHO classification5, we observed that, for the isocitrate dehydrogenase (IDH1/2) wild-type (WT) rGBM subgroup (n = 32 patients, 33 interventions), the mOS was 10.9 months (95% CI = 6.9–14.4 months), whereas, for the subgroup with recurrent IDHmutant (IDHmut) anaplastic astrocytoma (rAA; grade 3 or 4) (n = 4), the mOS was 5.4 months (95% CI = 2.6–∞ months) and, for the recurrent anaplastic oligodendroglioma (IDHmut; 1p/19q co-deleted), the mOS was 39.9 months (95% CI = 39.9–∞ months) (n = 5) (Fig. 1b). Progression-free survival times for the entire cohort and the cohort divided by the three rHGG diagnostic groups are shown in Extended Data Fig. 3a,b, respectively, and the clinical course of treated patients is shown in Extended Data Fig. 3c,d. Note that, in the swimmer plots, the timepoint of post-injection tumour resection is illustrated by a coloured triangle, with most additional antitumour therapies administered after resection. Full patient treatment histories have been included in Supplementary Table 1. Examples of significant clinical and radiographic responses are illustrated in Extended Data Fig. 4, including a response in a multifocal/multicentric rGBM.
Clinical trials of oncolytic-virus therapy in cancer have not shown that viral serology predicts response19,31. We checked whether HSV1 serology or seroconversion predicted survival in our study. In total, 14 out of 41 patients were seronegative for HSV1 before CAN-3110 treatment, with 4 out of 14 patients seroconverting after (Extended Data Table 3). Given the impact of IDHmut on survival32 and the small number of IDHmutpatients in the study, we focused analyses on the patients with IDHWT rGBM. Notably, HSV1 seropositivity both before and after treatment was associated with significantly longer survival after treatment (P = 0.009 and P = 0.007, respectively) (Extended Data Fig. 5a). In a survival analysis, HSV1-seropositive patients lived a median of 14.2 months (95% CI = 9.5–15.7 months) versus only 7.8 months (95% CI = 3.0–∞ months) for seronegative patients (P = 0.007, likelihood ratio test; Fig. 1c). By contrast, HSV2 serology was not associated with survival (P = 0.9, likelihood ratio test; Fig. 1d). Similarly, the trend towards longer survival for HSV1-seropositive patients was observed in the small number of patients with IDHmutrAA (Extended Data Fig. 5b). Cox proportional hazard analyses in IDHWT rGBMs validated pre-CAN-3110 positive HSV1 serology as a highly significant independent predictor of survival (Fig. 1e). As previously reported, age and tumour volume were also independent survival predictors33,34. These results therefore suggest the importance of an immunological mechanism for the response of patients with IDHWT rGBM to CAN-3110 therapy.
CAN-3110 increases T cells in tumours
There has been understandable reluctance to routinely collect rHGGs/rGBMs after an experimental therapy as it requires a surgical procedure. Even post-mortem examinations are rarely performed. To determine whether CAN-3110 induced a significant increase in lymphocytes in this lymphocyte-depleted tumour2, we endeavoured to recover as many post-treatment tumours as feasible either by re-resections at suspected progression and/or by post-mortem. Paired tumours from before and various timepoints after CAN-3110 treatment were analysed for a majority of separate rHGGs/rGBMs from patients after CAN-3110 treatment (Supplementary Table 2a–c and Supplementary Methods). In total, all analysed (except one) tumour pairs retained immunohistochemical expression for nestin and nectin-1, one of the major HSV receptors in cells35, both before and after injection (one tumour pair had insufficient material for pre-injection immunohistochemistry analysis) (Extended Data Fig. 6a,b and Supplementary Table 2b). Histological and immunohistochemical analyses showed increases in CD8+ and CD4+ tumour-infiltrating lymphocytes (TILs) in most paired tumours after CAN-3110 treatment (Extended Data Fig. 6c and Supplementary Table 2b). TILs could be visualized in a perivascular distribution, as well as with diffusely scattered cells and occasional clusters throughout the tumour (Extended Data Fig. 6d) and surrounding large areas of tumour necrosis (Extended Data Fig. 6e). Quantitative analyses showed a significant increase in CD4+ (P = 0.00085) and in CD8+ (P = 0.0034) TILs in most analysed paired tumours after CAN-3110 treatment (Fig. 2a and Supplementary Table 2c). There was a non-significant trend in CD20+ B cell increases in almost half of post-treatment samples. The most significant increases in CD8+ and CD4+ T cells were adjacent to perinecrotic areas that were possibly due to CAN-3110 cytotoxicity (Fig. 2b). The observed post-treatment increases in CD8+ and CD4+ T cells were significantly correlated with post-treatment survival in IDHWT rGBMs, but only in HSV1-seropositive patients (r = 0.58, P = 0.017 (CD8+) and r = 0.57, P = 0.026 (CD4+); Fig. 2c). Importantly, the overall quantitative assessments of CD8+, CD4+ and CD20+ TILs used in this analysis were not significantly confounded by the time of tissue collection (Extended Data Fig. 7a–c). Furthermore, longitudinal analyses of patient immune counts over time showed a non-significant trend towards a time-dependent decrease in CD8+ T cell numbers (albeit, without much change in CD4+ or B cells) over several months in HSV1-seronegative patients (Kruskal–Wallis test, P = 0.16; Extended Data Fig. 7d,e) more so than in HSV1-seropositive patients (P = 0.45), suggesting that the immune response induced by CAN-3110 may be durable over long periods of time in the latter. Multiplex immunofluorescence analysis in two of the analysed patients also showed CD68+ macrophage populations (specifically CD68+CD163+ myeloid cells expressing PD-L1) after CAN-3110 treatment, particularly in perinecrotic tumour regions (Extended Data Fig. 7f–i). These results therefore indicate that CAN-3110 induced an increase in TILs that was associated with longer survival in HSV1-seropositive patients but not in HSV1-seronegative patients.
Persistence is linked to seronegativity
It has been rare to find oncolytic viruses in injected tumours and, even when observed, persistence is limited to a few weeks21. We examined whether the observed immune infiltrates were associated with oHSV persistence in injected tumours. In 12 out of 29 tumours, oHSV antigen was present even several months after CAN-3110 injection (with the longest at 801 days) (Fig. 3a and Supplementary Table 2c). Importantly, in one case of multicentric GBM, a non-injected temporal lesion analysed 8 months after CAN-3110 injection showed positivity for HSV antigen in the absence of antigen detection in the original injected lesion (Fig. 3b). PCR was used to confirm the presence of CAN-3110-specific viral DNA, indicating probable ongoing replication, and spread from the injected lesion to the non-injected tumour (Extended Data Fig. 8). Coupled with the previous findings, these results showed that there was prolonged persistence of CAN-3110 in some patients, with increased CD4+ and CD8+ T cells in injected rHGGs in most participants and evidence of ongoing replication even in a tumour that was not initially injected in a patient with multicentric rGBM.
We examined whether the prolonged persistence of CAN-3110 in injected tumours was associated with HSV1 serological status. Indeed, oHSV persistence was significantly correlated with the absence of HSV1 seropositivity either before or after CAN-3110 treatment (Fig. 3c,d). These findings suggested that oHSV persistence in injected rHGGs/rGBMs may have been due to absence of a robust anti-HSV1 immune response. Coupled with the extended survival for patients with positive HSV1 serology (Fig. 1c), this suggests that tumour clearance of CAN-3110 characterized patients with an improved survival response to CAN-3110.
T cell metrics are linked to survival
The previous data (Fig. 2c) showed that CAN-3110 elicited an increased number of TILs in post-treatment samples that correlated with patient survival in the HSV1-seropositive patients. To further validate this finding, we examined whether survival was also correlated with changes in T cell clonotype metrics in tumour and/or peripheral blood mononuclear cells (PBMCs). Again, we focused the analyses on the IDHWT rGBM population: out of the 29 paired rHGGs/rGBMs, 21 were IDHWT rGBMs (corresponding to 20 patients). T cell receptor β chain (TCRβ) DNA sequencing (DNA-seq) was performed on tumours and corresponding PBMCs collected at various timepoints after injection (range, 7–349 days). These data were used to calculate changes in the T cell fraction and metrics of TCRβ diversity (productive entropy and productive Simpson clonality; Supplementary Methods). Again, these metrics were not significantly confounded by the collection timepoint (Extended Data Fig. 9a). We found that changes in the tumour T cell fraction (a measure of T cell frequency) after CAN-3110 treatment were positively correlated with prolonged post-treatment survival both in tumours of all of the patients and in tumours of the patients who were HSV1 seropositive (Fig. 4a and Extended Data Fig. 9b). Increased tumour TCRβ diversity (increased entropy/decreased clonality) was associated with prolonged post-treatment survival both in tumours of all of the patients and in tumours of patients who were HSV1 seropositive (Fig. 4b and Extended Data Fig. 9c,d). The same findings were observed for PBMCs (Fig. 4c,d and Extended Data Fig. 9e), suggesting that evolution of a polyclonal T cell response was correlated with survival. Notably, the association between HSV1 serology status and survival was maintained in the subset of patients with IDHWT rGBM for which TCRβ sequencing data were available (Extended Data Fig. 9f). Tumours from patients positive for HSV1 had nominally higher productive entropy (that is, higher TCRβ rearrangement diversity) compared with those from patients negative for HSV1 after (P = 0.070) but not before (P = 0.65) CAN-3110 treatment (Extended Data Fig. 9g), suggesting that TCRβ diversity after CAN-3110 treatment was influenced by positive HSV1 serological status.
We also performed bulk RNA-seq analysis of a subset of IDHWT rGBMs for which tumours were frozen (to obtain good-quality RNA) and identified transcripts that possessed a V(D)J junction (indicating a T or B cell receptor transcript). The total number of pre-treatment V(D)J transcripts was significantly correlated with post-treatment survival, with a trend towards significance with total post-treatment V(D)J transcript counts (Extended Data Fig. 9h,i), whereas the numbers of unique V(D)J transcripts both before and after treatment were significantly correlated with survival (Extended Data Fig. 9j,k), further validating the association between TCR abundance/diversity and post-treatment survival.
Specific public T cells are linked to survival
We next examined whether there were specific T cell clonotypes that were associated with participant response to therapy. To do this, we focused on public T cell clonotypes36, shared among the 21 IDHWT rGBMs for which we had TCRβ sequencing data. As expected, public TCRβs between patients were relatively rare in PBMCs and even more so in tumours (Extended Data Fig. 10a–f and Supplementary Methods). We found 55 public TCRβ sequences in 21 paired PBMC samples that we could analyse. There were highly significant changes in the frequency of two public PBMC T cell clones that were significantly associated with survival after treatment with CAN-3110: CASSLGGNTEAFF37,38 (Extended Data Fig. 10g; false-discovery rate (FDR) = 0.0035) and CASSSSTDTQYF39 ((Extended Data Fig. 10h; FDR = 0.018). Taken in conjunction, these findings show that survivorship after CAN-3110 treatment in the studied patients was significantly correlated with overall changes in T cell clonotype metrics and changes in the frequency of at least two specific public T cell clonotypes in PBMCs.
Changes in T cell repertoire
Given the little overlap (very few public TCRs) in TIL-specific TCR clonotypes between patients (Extended Data Fig. 10), the relationship between survival after CAN-3110 treatment and TCR clonotype frequency changes could not be meaningfully analysed in TILs. There has been recent interest in analyses of tumour/PBMC T cell clonal repertoire changes as a function of oncologic immunotherapy40. Similarly, we sought to determine whether the tumour/PBMC T cell clonal repertoire changed after treatment with CAN-3110. We found 63 TCRs that were significantly (FDR ≤ 0.05) expanded or depleted in TILs of 11 of the analysed patients with IDHWT rGBM (Supplementary Table 3). If we looked at TCRs that concordantly changed in TILs and PBMCs, four TCRs significantly (FDR ≤ 0.05) expanded and five TCRs were significantly depleted in both TILs and PBMCs (Extended Data Fig. 11a). Of the four expanded TCRs common between TILs and PBMCs, three were from a single patient—patient 021—who was an exceptional responder after CAN-3110 treatment and remained radiologically tumour free for more than 2 years after CAN-3110 treatment before dying due to a non-GBM-related event (Extended Data Fig. 4b and Supplementary Video 1). Notably, all TCRs that concordantly expanded/depleted in both TILs and PBMCs were in longer-surviving patients (Extended Data Fig. 11b), suggesting that defined and concordant PMBC/TIL T cell clonal repertoire changes denoted responses after CAN-3110 treatment. In one participant (previously discussed in Fig. 3b and Extended Data Fig. 8) who remained HSV1 seronegative throughout the trial and was therefore unlikely to have T cell reactivity against HSV1, there were four expanding emergent T cell clonotypes (Extended Data Fig. 11c). This suggested that these were unlikely to be reactive against CAN-3110. When assessing V(D)J gene usage, we also identified a correlation between post-treatment TCRBV09-01*01 (refs. 41,42) usage and survival in HSV1-seropositive patients (Extended Data Fig. 11d; Pearson’s r = 0.00019, FDR = 0.0095). Taken in conjunction, the analyses of T cell clonotypes in tumours revealed that longer-term survivors showed concordance between TIL and PBMC expansion, suggesting that there were alterations in the T cell repertoire after CAN-3110 treatment in the patients who survived for longer. In at least one participant, there was suggestive evidence that tumour TCR expansion was unlikely to be against CAN-3110.
Tumour immune signatures are linked to survival
We next queried RNA transcriptomic signatures in paired pre- and post-treatment frozen tumours (to maximize isolation of high-quality RNA) from 14 IDHWTrGBMs (13 patients, 14 interventions). Notably, associations between post-treatment immune signatures and survival were stronger when analysing samples from only HSV1-seropositive patients compared with when analysing samples from all patients (Fig. 5a–c and Extended Data Figs. 12 and 13). In fact, analysis in HSV1-seropositive patients showed 13 post-treatment immune signatures associated with survival (Fig. 5b,c and Extended Data Fig. 13b), whereas, when analyses were conducted with all patients (HSV1 seronegative and seropositive), there were only 7 post-treatment signatures associated with survival (Fig. 5b and Extended Data Fig. 12b). Notably, most of the immune signatures in HSV1-seropositive patients became associated with survival only after treatment with CAN-3110 (Fig. 5c). The time to tumour collection after treatment did not influence the post-treatment signature analyses (Extended Data Fig. 12c). When considered together with other data from this study (Fig. 5d), these results demonstrate that CAN-3110 instigates a highly inflammatory and immunologically activated tumour microenvironment in HSV1 serologically positive patients that persists beyond detectable HSV1 antigen and is significantly correlated with post-treatment survival in a way that is not true of the pretreatment tumour immune state.
Discussion
In this first-in-human clinical trial of CAN-3110, HSV meningitis or encephalitis was not seen, despite ongoing CAN-3110 persistence/replication for several months and maintenance of the ICP34.5 neurovirulence gene. All inflammatory responses remained confined to injected tumours and were not detected in the surrounding brain tissue. This was true in HSV-seropositive and HSV-seronegative patients. Overall, CAN-3110 was well tolerated without dose-limiting toxicities.
A major challenge faced by solid tumour immunotherapy is to create a microenvironment that is favourable for an efficient immune response against cancer cells43. CD8+ cytotoxic and CD4+ helper T cells are important by expressing effector programs against tumour antigens. More recently, public (for example, the same TCR sequence is shared between different individuals) T cell clones, some of which recognize shared viral antigens, have also been shown to traffic into tumours, and their function in cancer immunity is a subject of debate36. In this trial, we analysed a large majority of paired pre- and post-CAN-3110 rHGG/rGBM tumours, with corresponding longitudinal PBMCs to show that (1) pre-existing HSV1-positive serology correlated with individuals who survived the longest after treatment with CAN-3110; (2) CAN-3110 persisted in injected tumours, with almost half of assayed rHGGs still positive even months after a single timepoint injection, but persistence was significantly associated with negative HSV1 serology; and (3) CAN-3110 led to quantitative increases in TILs in a large majority of assayed tumours. Furthermore, we showed for the subpopulation with IDHWT rGBM, for whom there were available paired tumour samples, (4) improved patient survival was correlated with changes in T cell clonotype metrics (elevated T cell clone frequency, increased TCRβ rearrangement diversity, decreased clonality in post-injection versus pre-injection tumours, and transcripts associated with immunological effector programs, particularly in the individuals seropositive for HSV1); and (5) there were changes in specific public peripheral TCR clonotypes significantly associated with survival after CAN-3110 treatment. Taken together, positive HSV1 serology with the observed changes in T cell clonotypes, including public ones, results in a more efficacious immune response, characterizing individuals whose immune system is more ‘fit’ and who can mount a more effective antiviral and possibly antitumour immune response. Note that two of the longest survivors were treated with immune-checkpoint inhibition after their injected tumours were resected (see the swimmer plots of participant 019 and 021 in Extended Data Fig. 3c,d), based on the post-injection finding of extensive TILs. We speculate that CAN-3110 inflamed the TME, possibly improving the efficacy of immune-checkpoint inhibition therapy.
The finding that positive HSV1 serology before or after CAN-3110 treatment was a highly significant independent predictor of response was unexpected based on previously reported trials of other oHSVs16,17,19,31. A recent study showed no correlation between HSV1 serology in humans with GBM and survival44. We speculate that this finding may be specific to oncolytic viruses, based on the capacity of each oncolytic virus to replicate, persist and stimulate an innate and adaptive immune response. It may also be a factor related to sample size, at least for the brain tumour trials, as our trial had more participants. Note that the 22 participants (23 interventions) with IDHWT rGBM who were serologically positive for HSV1 before treatment with CAN-3110 had a mOS of 14.2 months (95% CI = 9.5–15.7 months; Fig. 1c), which is higher than the historical mOS of 6–9 months. Further prospective validation of this discovery in the next phase of planned trials will determine whether HSV1 serology can be used as a selection criterion for the likelihood of response.
The observation that CAN-3110 was immunohistochemically detected in almost half of the injected tumours several months (and even years in some patients) and even in one uninjected tumour suggests ongoing replication of the agent. Other oncolytic viruses, such as ICP34.5-defective oHSV, have rarely been found in injected human tumours, particularly after several weeks17,19–21,31,45–47, suggesting that CAN-3110 expression of ICP34.5 may enable persistence. We speculate that this persistence may increase infiltration of virus-specific TCR clones that could initially function in antitumour immunity in a bystander manner36, but could also begin to stimulate T cell responses against tumour antigen. Mouse brain tumour models do show that tumour infiltration of T cells against both tumour and viral antigens correlate with survival48. The significant association of HSV1 seropositivity with the absence of CAN-3110 antigen and transcripts in tumours after injection suggests that an initial humoral and probably adaptive antiviral immune response led to an improved antitumour response based on the survival data and on the finding that there were still increased CD8+ and CD4+ T cells and increased immunological transcriptional programs in tumours despite absent CAN-3110 in the longer-surviving patients (Fig. 6). Identification of the expansion of emergent TCRs, such as those in patient 014 who was seronegative for HSV1 before and after CAN-3110 treatment, possibly suggest that oHSV therapy indeed promotes epitope spreading49, enabling expansion of T cell clones against tumour antigens. Future extensive studies determining whether the TCRs that we discovered in injected tumours react to viral versus tumour antigens are underway (data not shown).
In summary, single-timepoint intralesional injection of rHGG/rGBM with CAN-3110 enriches the tumour microenvironment with TILs, inducing defined changes in peripheral and tumour T cell repertoires and tumour transcriptomic signatures. These changes are particularly evident in patients who are seropositive for HSV1 and are associated with improved survival in this otherwise therapy-refractory cancer. These findings therefore provide human immunological and biological evidence supporting intralesional oncolytic modalities to convert the immunosuppressive TME characteristic of many solid cancers into a TME that is more favourable to immunologic rejection of the tumour. We are now set to determine whether multiple-timepoint injections lead to further improvements in this therapy (ClinicalTrials.gov: NCT03152318).
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Online content
Any methods, additional references, Nature Portfolio reporting summaries, source data, extended data, supplementary information, acknowledgements, peer review information; details of author contributions and competing interests; and statements of data and code availability are available at 10.1038/s41586-023-06623-2.
Supplementary information
Source data
Acknowledgements
We thank all of the patients and families who participated in the trial, particularly participant 021 and her family. We acknowledge assistance from S. Ventz, G. Park, L. M. Seften, K. Hoe Chow, A. Shetty, W. Pisano, E. Lapinskas and F. Watkinson; D. Hamm and B. Banbury for assistance in TCR analyses; and N. Caffo, J. Dwyer, D. Lane and H. White for assistance.
Extended data figures and tables
Author contributions
Conceptualization: E.A.C. Methodology: A.L.L., I.H.S., A.M.L., H.N., J.K.W., A. Santos, N. Masud, G.F., X.M., A.S.Y., J.G., A.Z., J.D.B., E. Torio, H.I., J.L., N. Shono, M.O.N., P.H., R.P., H.S., B.S., W.L.B., P.P., E.C., S.W.M., M.C.P., Y.Z., A.C., S.J.R., K.K., E. Tikhonova, N. Miheecheva, D. Tabakov, N. Shin, A.G., A. Shumskiy, F.F., E.A.-C., L.K.A., J.W., D. Krisky, A.M., C.M., P.P.T., F.B., D. Kovarsky, I.T., M.L.S., K.W.W., K.L., D.A.R. and E.A.C. Investigation: A.L.L., I.H.S., A.M.L., H.N., J.K.W., A. Santos, N. Masud, G.F., X.M., A.S.Y., J.G., A.Z., J.D.B., E. Torio, H.I., J.L., N. Shono, M.O.N., D. Triggs, P.H., R.P., H.S., B.S., W.L.B., P.P., S.W.M., M.C.P., Y.Z., A.C., S.J.R., P.Y.W., E.Q.L., L.N., U.C., L.N.G.C., S.D.D., T.B., K.K., E. Tikhonova, N. Miheecheva, D. Tabakov, N. Shin, A.G., A. Shumskiy, F.F., E.A.-C., L.K.A., D. Krisky, J.W., A.M., C.M., P.P.T., F.B., D. Kovarsky, I.T., M.L.S., K.W.W., K.L., D.A.R. and E.A.C. Visualization: A.L.L., I.H.S., A.M.L., H.N., A. Santos, G.F., X.M., N. Shono, S.W.M., M.C.P., E. Tikhonova, N. Miheecheva, D. Tabakov, N. Shin, A.G., A. Shumskiy, F.F., F.B., K.L. and E.A.C. Funding acquisition: A.L.L., J.K.W., N. Shono, E.A.-C., P.P.T., M.L.S., K.W.W., K.L., D.A.R. and E.A.C. Project administration: H.N., S.J.R., P.Y.W., T.B., N. Shin, E.A.-C., P.P.T., F.B., I.T., M.L.S., K.W.W., K.L., D.A.R. and E.A.C. Supervision: I.H.S., H.N., G.F., X.M., S.J.R., P.Y.W., T.B., N. Shin, P.P.T., F.B., E.A.-C., L.K.A., M.L.S., K.W.W., I.T., K.L., D.A.R. and E.A.C. Writing—original draft: A.L.L. and E.A.C. Writing—review and editing: A.L.L., I.H.S., A.M.L., H.N., J.K.W., G.F., X.M., J.D.B., M.C.P., P.Y.W., L.N.G.C., N. Shin, E.A.-C., L.K.A., P.P.T., F.B., D. Kovarsky, M.L.S., K.W.W., D.A.R. and E.A.C.
Peer review
Peer review information
Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.
Data availability
Patient responses, demographic information and safety outcomes, as well IHC quantifications and RNA-seq gene signature scores are available within the Article and its Supplementary Information. Raw RNA-seq and TCRβ DNA-seq files have been deposited in a controlled-access repository in the Database of Genotypes and Phenotypes (http://www.ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs003378.v1.p1). Source data are provided with this paper.
Code availability
Custom code used to perform analyses in this study have been deposited at OSF (10.17605/OSF.IO/YBCG7).
Competing interests
J.D.B. has an equity position in Treovir, an oHSV clinical stage company and is a member of the POCKiT Diagnostics board of scientific advisors; and has a provisional patent (application number 63/273,577) entitled ‘Methods and formulations related to the intrathecal delivery of oncolytic viruses’. W.L.B. consulted for Stryker. P.P. is cofounder and member of the board of directors of Ternalys Therapeutics; he is also named as an inventor on patents related to non-coding RNA technology. P.Y.W. received research support from AstraZeneca/Medimmune, Beigene, Celgene, Chimerix, Eli Lily, Genentech/Roche, Kazia, MediciNova, Merck, Novartis, Nuvation Bio, Puma, Servier, Vascular Biogenics and VBI Vaccines and served on advisory boards for AstraZeneca, Bayer, Black Diamond, Boehringer Ingelheim, Boston Pharmaceuticals, Celularity, Chimerix, Genenta, GlaxoSmithKline, Karyopharm, Merck, Mundipharma, Novartis, Novocure, Nuvation Bio, Prelude Therapeutics, Sapience, Servier, Sagimet, Vascular Biogenics and VBI Vaccines, and on data safety monitoring committees for Day One Bio and Novocure. L.N. serves as a consultant for Ono and Brave Bio. L.N.G.C. received research support from Merck (to the Dana-Farber Cancer Institute); he also has received research support from the NIH, the American Society of Clinical Oncology and the Robert Wood-Johnson Foundation. E.Q.L. receives royalties from Wolter Kluwer (Up to Date) and consulting fees from GCAR. T.B. receives clinical trial support from ONO Pharmaceuticals, publishing royalties from UpToDate and Oxford University Press. S.J.R. receives research support from Bristol Myers-Squibb and KITE/Gilead; and is on the scientific advisory board for Immunitas Therapeutics. K.K. is employed by and owns equity in Clearpoint. E. Tikhonova, N. Miheecheva, D. Tabakov, N. Shin, A.G., A. Shumskiy and F.F. are employed by BostonGene and have equity options in BostonGene. E.A.-C. is a founder, board member of and holds equity in Candel Therapeutics. L.K.A. is co-founder and holds equity in Candel Therapeutics. D. Krisky, J.W., A.M., C.M., P.P.T. and F.B. are employees of and hold equity in Candel Therapeutics. I.T. is an advisory board member of Immunitas Therapeutics. M.L.S. and K.W.W. are equity holders, scientific co-founders and advisory board members of Immunitas Therapeutics. D.A.R. is an advisor to Agios, AnHeart Therapeutics, Avita Biomedical, Blue Rock Therapeutics, Bristol Myers Squibb, Boston Biomedical, CureVac, Del Mar Pharma, DNAtrix, Hoffman-LaRoche, Imvax, Janssen, Kiyatec, Medicenna Therapeutics, Neuvogen, Novartis, Novocure, Pyramid, Sumitomo Dainippon Pharma, Vivacitas Oncology and Y-mabs Therapeutics. E.A.C. is an advisor to Amacathera, Bionaut Labs, Genenta, Insightec, DNAtrix, Seneca Therapeutics and Theravir; he has equity options in Bionaut Laboratories, DNAtrix, Immunomic Therapeutics, Seneca Therapeutics and Ternalys Therapeutics; he is co-founder and on the board of directors of Ternalys Therapeutics. Patents related to oHSV and CAN-3110 are under the possession of Brigham and Women’s Hospital with E.A.C. and H.N. named as co-inventors. These patents have been licensed to Candel Therapeutics. Present and future milestone license fees and future royalty fees are distributed to Brigham and Women’s Hospital from Candel. The other authors declare no competing interests.
Footnotes
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
These authors contributed equally: Alexander L. Ling, Isaac H. Solomon, Ana Montalvo Landivar, Hiroshi Nakashima
Change history
11/3/2023
An amendment to the underlying article code was made to enable an author name to appear correctly in PubMed. In addition, a hyphen was missing in the data availability link, which has now been updated.
Extended data
is available for this paper at 10.1038/s41586-023-06623-2.
Supplementary information
The online version contains supplementary material available at 10.1038/s41586-023-06623-2.
References
- 1.Chuntova, P. et al. Unique challenges for glioblastoma immunotherapy-discussions across neuro-oncology and non-neuro-oncology experts in cancer immunology. Meeting report from the 2019 SNO Immuno-Oncology Think Tank. Neuro Oncol.23, 356–375 (2021). 10.1093/neuonc/noaa277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Thorsson, V. et al. The immune landscape of cancer. Immunity48, 812–830 (2018). 10.1016/j.immuni.2018.03.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Chiocca, E. A., Nakashima, H., Kasai, K., Fernandez, S. A. & Oglesbee, M. Preclinical toxicology of rQNestin34.5v.2: an oncolytic herpes virus with transcriptional regulation of the ICP34.5 neurovirulence gene. Mol. Ther. Methods Clin. Dev.17, 871–893 (2020). 10.1016/j.omtm.2020.03.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kambara, H., Okano, H., Chiocca, E. A. & Saeki, Y. An oncolytic HSV-1 mutant expressing ICP34.5 under control of a nestin promoter increases survival of animals even when symptomatic from a brain tumor. Cancer Res.65, 2832–2839 (2005). 10.1158/0008-5472.CAN-04-3227 [DOI] [PubMed] [Google Scholar]
- 5.Louis, D. N. et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol.23, 1231–1251 (2021). 10.1093/neuonc/noab106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wen, P. Y. & Packer, R. J. The 2021 WHO classification of tumors of the central nervous system: clinical implications. Neuro Oncol.23, 1215–1217 (2021). 10.1093/neuonc/noab120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Vargas Lopez, A. J. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol.23, 502–503 (2021). 10.1093/neuonc/noaa287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Taslimi, S., Ye, V. C., Wen, P. Y. & Zadeh, G. Lessons learned from contemporary glioblastoma randomized clinical trials through systematic review and network meta-analysis: part 2 recurrent glioblastoma. Neurooncol. Adv.3, vdab029 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Macedo, N., Miller, D. M., Haq, R. & Kaufman, H. L. Clinical landscape of oncolytic virus research in 2020. J. Immunother. Cancer8, e001486 (2020). 10.1136/jitc-2020-001486 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Martin, N. T. & Bell, J. C. Oncolytic virus combination therapy: killing one bird with two stones. Mol. Ther.26, 1414–1422 (2018). 10.1016/j.ymthe.2018.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Peruzzi, P. & Chiocca, E. A. Viruses in cancer therapy—from benchwarmers to quarterbacks. Nat. Rev. Clin. Oncol.15, 657–658 (2018). 10.1038/s41571-018-0077-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Russell, L. & Peng, K. W. The emerging role of oncolytic virus therapy against cancer. Chin. Clin. Oncol.7, 16 (2018). 10.21037/cco.2018.04.04 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Totsch, S. K. et al. Oncolytic herpes simplex virus immunotherapy for brain tumors: current pitfalls and emerging strategies to overcome therapeutic resistance. Oncogene38, 6159–6171 (2019). 10.1038/s41388-019-0870-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Harrington, K. J. et al. Clinical development of talimogene laherparepvec (T-VEC): a modified herpes simplex virus type-1-derived oncolytic immunotherapy. Expert Rev. Anticancer Ther.15, 1389–1403 (2015). 10.1586/14737140.2015.1115725 [DOI] [PubMed] [Google Scholar]
- 15.Christie, J. D. & Chiocca, E. A. Treat and repeat: oncolytic virus therapy for brain cancer. Nat. Med.28, 1540–1542 (2022). 10.1038/s41591-022-01901-4 [DOI] [PubMed] [Google Scholar]
- 16.Todo, T., Ino, Y., Ohtsu, H., Shibahara, J. & Tanaka, M. A phase I/II study of triple-mutated oncolytic herpes virus G47 in patients with progressive glioblastoma. Nat. Commun.13, 4119 (2022). 10.1038/s41467-022-31262-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Todo, T. et al. Intratumoral oncolytic herpes virus G47 for residual or recurrent glioblastoma: a phase 2 trial. Nat. Med.28, 1630–1639 (2022). 10.1038/s41591-022-01897-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Desjardins, A. et al. Recurrent glioblastoma treated with recombinant poliovirus. N. Engl. J. Med.379, 150–161 (2018). 10.1056/NEJMoa1716435 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gallego Perez-Larraya, J. et al. Oncolytic DNX-2401 virus for pediatric diffuse intrinsic pontine glioma. N. Engl. J. Med.386, 2471–2481 (2022). 10.1056/NEJMoa2202028 [DOI] [PubMed] [Google Scholar]
- 20.Friedman, G. K. et al. Oncolytic HSV-1 G207 immunovirotherapy for pediatric high-grade gliomas. N. Engl. J. Med.384, 1613–1622 (2021). 10.1056/NEJMoa2024947 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lang, F. F. et al. Phase I study of DNX-2401 (Delta-24-RGD) oncolytic adenovirus: replication and immunotherapeutic effects in recurrent malignant glioma. J. Clin. Oncol.36, 1419–1427 (2018). 10.1200/JCO.2017.75.8219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Fares, J. et al. Neural stem cell delivery of an oncolytic adenovirus in newly diagnosed malignant glioma: a first-in-human, phase 1, dose-escalation trial. Lancet Oncol.22, 1103–1114 (2021). 10.1016/S1470-2045(21)00245-X [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.van Putten, E. H. P. et al. Convection enhanced delivery of the oncolytic adenovirus Delta24-RGD in patients with recurrent GBM: a phase I clinical trial including correlative studies. Clin. Cancer Res.28, 1572–1585 (2022). 10.1158/1078-0432.CCR-21-3324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kanai, R. et al. Effect of γ34.5 deletions on oncolytic herpes simplex virus activity in brain tumors. J. Virol.86, 4420–4431 (2012). 10.1128/JVI.00017-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Peters, C. et al. Restriction of replication of oncolytic herpes simplex virus with a deletion of γ34.5 in glioblastoma stem-like cells. J. Virol.10.1128/JVI.00246-18 (2018). [DOI] [PMC free article] [PubMed]
- 26.Orvedahl, A. et al. HSV-1 ICP34.5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell Host Microbe1, 23–35 (2007). 10.1016/j.chom.2006.12.001 [DOI] [PubMed] [Google Scholar]
- 27.Aghi, M., Visted, T., Depinho, R. A. & Chiocca, E. A. Oncolytic herpes virus with defective ICP6 specifically replicates in quiescent cells with homozygous genetic mutations in p16. Oncogene27, 4249–4254 (2008). 10.1038/onc.2008.53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Fyllingen, E. H. et al. Survival of glioblastoma in relation to tumor location: a statistical tumor atlas of a population-based cohort. Acta Neurochir.163, 1895–1905 (2021). 10.1007/s00701-021-04802-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lasocki, A., Gaillard, F., Tacey, M., Drummond, K. & Stuckey, S. Multifocal and multicentric glioblastoma: Improved characterisation with FLAIR imaging and prognostic implications. J. Clin. Neurosci.31, 92–98 (2016). 10.1016/j.jocn.2016.02.022 [DOI] [PubMed] [Google Scholar]
- 30.Li, Y. et al. A systematic review of multifocal and multicentric glioblastoma. J. Clin. Neurosci.83, 71–76 (2021). 10.1016/j.jocn.2020.11.025 [DOI] [PubMed] [Google Scholar]
- 31.Andtbacka, R. H. et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J. Clin. Oncol.33, 2780–2788 (2015). 10.1200/JCO.2014.58.3377 [DOI] [PubMed] [Google Scholar]
- 32.Yan, H. et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med.360, 765–773 (2009). 10.1056/NEJMoa0808710 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ohgaki, H. & Kleihues, P. Epidemiology and etiology of gliomas. Acta Neuropathol.109, 93–108 (2005). 10.1007/s00401-005-0991-y [DOI] [PubMed] [Google Scholar]
- 34.Keles, G. E., Lamborn, K. R., Chang, S. M., Prados, M. D. & Berger, M. S. Volume of residual disease as a predictor of outcome in adult patients with recurrent supratentorial glioblastomas multiforme who are undergoing chemotherapy. J. Neurosurg.100, 41–46 (2004). 10.3171/jns.2004.100.1.0041 [DOI] [PubMed] [Google Scholar]
- 35.Huang, Y. et al. Receptors and ligands for herpes simplex viruses: novel insights for drug targeting. Drug Discov. Today27, 185–195 (2022). 10.1016/j.drudis.2021.10.004 [DOI] [PubMed] [Google Scholar]
- 36.Meier, S. L., Satpathy, A. T. & Wells, D. K. Bystander T cells in cancer immunology and therapy. Nat. Cancer3, 143–155 (2022). 10.1038/s43018-022-00335-8 [DOI] [PubMed] [Google Scholar]
- 37.Massa, C. et al. Identification of patient-specific and tumor-shared T cell receptor sequences in renal cell carcinoma patients. Oncotarget8, 21212–21228 (2017). 10.18632/oncotarget.15064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Werner, L. et al. Alterations in T and B cell receptor repertoires patterns in patients with IL10 signaling defects and history of infantile-onset IBD. Front. Immunol.11, 109 (2020). 10.3389/fimmu.2020.00109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tickotsky, N., Sagiv, T., Prilusky, J., Shifrut, E. & Friedman, N. McPAS-TCR: a manually curated catalogue of pathology-associated T cell receptor sequences. Bioinformatics33, 2924–2929 (2017). 10.1093/bioinformatics/btx286 [DOI] [PubMed] [Google Scholar]
- 40.Luoma, A. M. et al. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell185, 2918–2935 (2022). 10.1016/j.cell.2022.06.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Held, K. et al. αβ T-cell receptors from multiple sclerosis brain lesions show MAIT cell-related features. Neurol. Neuroimmunol. Neuroinflamm.2, e107 (2015). 10.1212/NXI.0000000000000107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Savola, P. et al. Somatic mutations in clonally expanded cytotoxic T lymphocytes in patients with newly diagnosed rheumatoid arthritis. Nat. Commun.8, 15869 (2017). 10.1038/ncomms15869 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tomaszewski, W., Sanchez-Perez, L., Gajewski, T. F. & Sampson, J. H. Brain tumor microenvironment and host state: implications for immunotherapy. Clin. Cancer Res.25, 4202–4210 (2019). 10.1158/1078-0432.CCR-18-1627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Guerra, G. et al. Antibodies to varicella-zoster virus and three other herpesviruses and survival in adults with glioma. Neuro Oncol.10.1093/neuonc/noac283 (2023). [DOI] [PMC free article] [PubMed]
- 45.Harrow, S. et al. HSV1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: safety data and long-term survival. Gene Ther.11, 1648–1658 (2004). 10.1038/sj.gt.3302289 [DOI] [PubMed] [Google Scholar]
- 46.Markert, J. M. et al. Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM. Mol. Ther.17, 199–207 (2009). 10.1038/mt.2008.228 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ribas, A. et al. Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell170, 1109–1119 (2017). 10.1016/j.cell.2017.08.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Alayo, Q. A. et al. Glioblastoma infiltration of both tumor- and virus-antigen specific cytotoxic T cells correlates with experimental virotherapy responses. Sci. Rep.10, 5095 (2020). 10.1038/s41598-020-61736-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hu, Z. et al. Personal neoantigen vaccines induce persistent memory T cell responses and epitope spreading in patients with melanoma. Nat. Med.27, 515–525 (2021). 10.1038/s41591-020-01206-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Patient responses, demographic information and safety outcomes, as well IHC quantifications and RNA-seq gene signature scores are available within the Article and its Supplementary Information. Raw RNA-seq and TCRβ DNA-seq files have been deposited in a controlled-access repository in the Database of Genotypes and Phenotypes (http://www.ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs003378.v1.p1). Source data are provided with this paper.
Custom code used to perform analyses in this study have been deposited at OSF (10.17605/OSF.IO/YBCG7).