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 antigens9,10,11,12,13. 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 Japan15,16,17. Notably, several early-phase oncolytic-virus clinical trials for HGG have been published in recent high-profile literature17,18,19,20,21,22,23. 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.

Table 1 Total adverse events (grade 1 or 2) related to CAN-3110
Table 2 Serious adverse events (grade 3 or above) possibly, likely or definitely related to CAN-3110

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 survival28,29,30, 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.

Fig. 1: Survival data.
figure 1

a, Kaplan–Meier survival analysis of 41 patients with rHGG (42 interventions) after treatment with CAN-3110 (day 0). The shaded area shows the 95% CIs; the Kaplan–Meier estimate of survival probability is shown. Data maturity, October 2022. Median survival time (MST), 11.6 months (95% CI = 7.8–14.9 months). b, Kaplan–Meier survival analysis of patients with IDHWT rGBM (n = 32 patients, 33 interventions), IDHmutrAA (grades 3 and 4; n = 4 patients) and IDHmut rAO (grade 3; n = 5 patients). MST, 10.9 months (IDHWTrGBM; 95% CI = 6.9–14.4 months), 5.4 months (IDHmutrAA; 95% CI = 2.6–∞ months) and 39.9 months (IDHmut rAO; 95% CI = 39.9–∞ months). Hazard ratio (HR): IDHmutrAO, 0.07 (95% CI = 0.01–0.49, P = 0.0079, two-sided Cox proportional-hazard test); IDHmutrAA, 1.09 (95% CI = 0.38–3.16, P = 0.87, two-sided Cox proportional-hazard test). c, Kaplan–Meier survival analysis of 31 patients with IDHWT rGBM (32 interventions) by negative (n = 9) or positive (n = 22 patients, 23 interventions) HSV1 serological status after treatment with CAN-3110. MST, HSV1 positive, 14.2 months (95% CI = 9.5–15.7 months); and HSV1 negative, 7.8 months (95% CI = 3.0–∞ months). P = 0.007 (two-sided likelihood ratio test). d, Kaplan–Meier survival analysis of 31 patients with IDHWT rGBM (32 interventions) by negative (n = 24 patients, 25 interventions) or positive (n = 7) HSV2 serological status before treatment with CAN-3110. MST, HSV2 positive, 6.9 months (95% CI = 2.2–∞ months); and HSV2 negative, 11.8 months (95% CI = 8.3–14.5 months). P = 0.9 (two-sided likelihood ratio test). e, Cox proportional-hazard ratio multivariate analyses for independent predictors of survival in patients with IDHWT rGBM after treatment with CAN-3110. The error bars and values in parentheses show the 95% CIs. P values calculated using two-sided Cox proportional-hazard tests are shown on the right for each covariate. The unit of tumour volume is increments of 10 cm3. Partial MGMT promoter methylation was treated as unmethylated. For patients who were administered dexamethasone within the 30 days before or after CAN-3110 treatment, the median dose was 4 mg per day, and the median number of treated days during this time was 14.5 days. KPS, Karnofsky performance score. For ce, participant 045 was excluded due to non-GBM mortality. PFU, plaque-forming units.

Source Data

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.

Fig. 2: Neuropathologic analyses.
figure 2

a, Quantification of CD4+ and CD8+ T cells and CD20+ B cells from patients with available paired pre-treatment biopsies and post-treatment tumour samples distal from and/or directly adjacent to the virus injection site. n = 26 patients and 27 interventions (CD8+), and 24 patients and 25 interventions (CD4+ and CD20+). P values were calculated using two-sided Wilcoxon matched-pairs signed-rank tests. b, Quantification of CD8+, CD4+ and CD20+ cells in pre-treatment and post-treatment samples in tumour areas far from the CAN-3110 injection site versus tumour areas near to necrotic foci associated with CAN-3110 injection. For pre-treatment, post-treatment and perinecrotic areas, respectively, n patients (interventions) = 39 (40), 29 (30) and 6 (6) (CD8+); 37 (38), 29 (30) and 3 (CD4+); 36 (37), 29 (30) and 2 (2) (CD20+). c, Correlations between changes in immune counts and post-treatment survival for CD8+ (left), CD4+ (middle) and CD20+ (right) cells in IDHWT rGBMs. Pearson’s correlation coefficient r and P values (two-sided, based on t-distribution) are provided above each plot calculated either using all patients or using only patients who were HSV1 seropositive before or after treatment. When counts were available for multiple post-treatment timepoints for a patient, the timepoint with the highest number of CD4+CD8+ cells was chosen. Importantly, TIL counts were not significantly confounded by the collection timepoint (Extended Data Fig. 7a–c). Patient 045 was excluded from the analyses in c due to early non-GBM mortality. The box plots show the median (centre line), 25th and 75th percentiles (box limits) and up to 1.5× the interquartile range or to the minimum/maximum values (if <1.5 × interquartile range distance from the box) (whiskers).

Source Data

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.

Fig. 3: CAN-3110 persistence in injected rHGG/rGBM is associated with negative HSV1 serological status either before or after therapy.
figure 3

a, oHSV-positive immunohistochemistry (IHC) images from two participants. Top, magnetic resonance imaging (MRI) images before and 41 days after CAN-3110 injection (106 PFU) from patient 005. oHSV-positive immunohistochemistry was visualized in the large area of tumour necrosis. The area was also positive for oHSV DNA as determined using PCR and positive for ICP22 oHSV transcripts as determined using quantitative PCR with reverse transcription (RT–qPCR; data not shown). Bottom, MRI images from patient 028 before and 253 days after CAN-3110 injection (109 PFU). oHSV-positive immunohistochemistry images were visualized in the area of resected tumour necrosis; the positive status of ICP22 oHSV transcript was determined using RT–qPCR (data not shown). b, Participant 014 had multifocal GBMs in the left temporal and left occipital lobes. The left occipital lobe lesion was injected with 107 PFU of CAN-3110. Post-mortem analyses were performed 252 days after injection. Top left, MRI scan before post-mortem brain collection, with the necrotic injected occipital lesion, shown in the grossly necrotic lesion (top middle), confirmed by histological haematoxylin and eosin (H&E) staining (top right). The CAN-3110 non-injected temporal-lobe post-mortem gross section (bottom left) exhibited oHSV positivity (bottom middle) and dense infiltrates of CD8+ T cells (bottom right). Extended Data Fig. 8 shows that this oHSV-positive focus was CAN-3110 and not reactivated latent wild-type HSV1 from this patient who was otherwise seronegative for HSV throughout the trial. c, HSV1 pathology staining in tumour tissue from patients with rGBM/rHGG (n = 28 interventions, 27 patients) after CAN-3110 treatment relative to HSV1 serological status. d, The same data as in c, but with patients who were initially seropositive grouped with patients who seroconverted after treatment with CAN-3110. Focal/weak pathology staining was grouped with negative staining; and multifocal staining was grouped with positive staining. P values were calculated using two-sided Fisher’s exact tests. For a and b, scale bars, 100 µm.

Source Data

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.

Fig. 4: TCR clonotype analyses.
figure 4

a, The correlation between the change in tumour T cell fraction (after versus before CAN-3110 treatment) and survival after CAN-3110 treatment. The T cell fraction is the fraction of nucleated cells that are T cells on the basis of TCRβ DNA-seq analysis (see the ‘Definition of TCR based metrics’ section in the Supplementary Methods). b, The correlation between post-CAN-3110 tumour TCR productive entropy (Supplementary Methods) and survival. A higher entropy indicates a greater diversity of TCRβ rearrangements. n = 18 interventions and 17 patients. For a and b, three participants were excluded (two who survived longer than 1 year, and one who survived less than 1 year) with <200 ng of gDNA. n = 18 interventions and 17 patients. Extended Data Fig. 9b,c shows analyses with all patients, regardless of the amount of gDNA collected. c, The correlation between the change in PBMC TCR clonotype fraction (after versus before CAN-3110 treatment) and survival after CAN-3110 treatment. n = 21 interventions and 20 patients. For ac, Pearson’s r correlation coefficients and P values (two-sided, based on t-distribution) are shown above the plots. d, Kaplan–Meier survival analysis based on an increase (change > 0) or decrease (change < 0) in PBMC productive Simpson’s clonality (Supplementary Methods) after CAN-3110 treatment. HRincreased = 2.79 (95% CI = 1.08–7.21), P = 0.034 (two-sided Cox proportional-hazard test). Higher clonality indicates a lower diversity of TCRβ rearrangements.

Source Data

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.

Fig. 5: Survival correlation between immune transcript signature programs in HSV1-seronegative and HSV1-seropositive patients.
figure 5

A total of 13 paired IDHWTrGBMs with good-quality RNA was analysed by bulk RNA transcriptomics. Transcriptomic signatures for different biological programs were estimated for each sample, and these signatures were assessed for correlation with survival after CAN-3110 treatment either in all patients or only in patients who were HSV1 seropositive before or after CAN-3110 treatment. a, Example of two immune signatures (antitumour cytokine and T cell signatures) that are strongly correlated with survival after CAN-3110 treatment when analysed in HSV1 seropositive patients. Pearson’s r correlation coefficients and P values (two-sided, based on t-distribution) are shown above the plots. Importantly, these signatures did not appear to be significantly confounded by the tissue collection timepoint (Extended Data Fig. 12c). b, The change in Pearson’s correlation P (left) (two-sided, based on t-distribution) and r (right) values when correlations between post-treatment immune signatures and survival were performed in all patients (red points) or in only HSV1-seropositive patients (teal points). Only gene signatures that reached P ≤ 0.05 (dashed red line) in either analysis were plotted. DCs, dendritic cells; MDSCs, myeloid-derived suppressor cells; TH1, T helper 1. c, The change in Pearson’s correlation P (left) (two-sided, based on t-distribution) and r (right) values for pre-treatment (red points) and post-treatment (teal points) samples from HSV1-seropositive patients. This panel includes all of the analysed RNA-seq gene signatures. The dashed red line indicates P = 0.05. CAFs, cancer-associated fibroblasts; EMT, epithelial–mesenchymal transition; NK cells, natural killer cells; TAMM, tumour-associated monocyte/macrophage; Treg cells, regulatory T cells. d, Combined data for all of the patients in the study, including survival after CAN-3110 treatment, HSV1 serology, HSV1 tumour pathology, T cell fraction changes based on TCRβ DNA-seq, initial tumour volumes and bulk RNA-seq-based antitumour cytokine signature scores. The grey boxes indicate missing data. For b and c, HSV1 serology remained unchanged after CAN-3110 treatment for all of the patients, and one patient (045) was omitted from the analysis due to early non-GBM mortality.

Source Data

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 weeks17,19,20,21,31,45,46,47, 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).

Fig. 6: A model for CAN-3110 action as a function of HSV1 serology.
figure 6

In patients who are seropositive for HSV1, CAN-3110 elicits an initial augmented anti-HSV1 innate and T cell-mediated response (presumably by expansion and differentiation of memory into effector anti-HSV1 T cells) to clear the injected oHSV from tumours. This bystander T cell effect possibly mediates an effective antitumour effect by direct inflammation in the tumour and/or by stimulating ‘antigen spreading’ to also elicit T cell recognition of tumour antigens. In patients who are seronegative for HSV1, the absence of a rapid anti-HSV1 innate and T cell response leads to CAN-3110 replicative persistence with tumour growth overcoming viral-induced cytotoxicity and delayed immune activity against tumour antigens. The figure was generated using BioRender.com.

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.