Skip to main content

Neoadjuvant immunotherapy across cancers: meeting report from the Immunotherapy Bridge—December 1st–2nd, 2021

Abstract

After the success of immunotherapy in the treatment of advanced metastatic cancer, further evaluation in earlier settings, including high-risk, surgically-resectable disease is underway. Potential benefits of a neoadjuvant immunotherapeutic approach include presurgical tumor shrinkage, reduced surgical morbidity, early eradication of micrometastases and prevention of distant disease, and greater antigen-specific T cell response. For some cancers, pathologic response has been established as a surrogate measure for long-term outcomes, therefore offering the ability for early and objective assessment of treatment efficacy and the potential to inform and personalize adjuvant treatment clinical decision-making. Leveraging the neoadjuvant treatment setting offers the ability to deeply interrogate longitudinal tissue in order to gain translatable, pan-malignancy insights into response and mechanisms of resistance to immunotherapy. Neoadjuvant immunotherapy across cancers was a focus of discussion at the virtual Immunotherapy Bridge meeting (December 1–2, 2021). Clinical, biomarker, and pathologic insights from prostate, breast, colon, and non-small-cell lung cancers, melanoma and non-melanoma skin cancers were discussed and are summarized in this report.

Introduction

The development of immunotherapy has revolutionized treatment of metastatic cancer and improved survival for patients across many types of cancer. After demonstrating marked success in the treatment of advanced malignancies, further evaluation of immunotherapy in earlier settings, including high-risk, surgically-resectable disease has been under investigation. Earlier administration of therapies shown to be effective in a metastatic setting may offer further benefits and neoadjuvant therapy has an established role in several cancers. Notable benefits of a neoadjuvant approach include reduced surgical morbidity through tumor shrinkage, and the opportunity for early eradication of micrometastases and prevention of distant disease. Preclinical data suggest neoadjuvant checkpoint inhibition is superior to adjuvant checkpoint inhibition, with increased eradication of distant metastases, improved survival and greater antigen-specific T cell response following primary tumor resection in a murine breast cancer model [1]. For some cancers, pathologic response has been established as a surrogate measure for long-term outcomes, therefore offering the ability for early and objective assessment of treatment efficacy and the potential to inform and personalize adjuvant treatment clinical decision-making [2,3,4] Leveraging the neoadjuvant treatment setting offers the ability to deeply interrogate longitudinal tissue in order to gain translatable, pan-malignancy insights into response and mechanisms of resistance to immunotherapy.

Neoadjuvant immunotherapy across cancers was a focus of discussion at the virtual Immunotherapy Bridge meeting (December 1–2, 2021). Clinical, biomarker, and pathologic insights from prostate, breast, colon, and non-small-cell lung cancers (NSCLC), melanoma and non-melanoma skin cancers were discussed and are summarized in this report.

From the clinic to the laboratory: investigating mechanism of resistance and response to immune checkpoint therapy

Large numbers of clinical immunotherapy trials are ongoing, which include patients who are polymorphic and with heterogeneous disease. These studies can generate hypotheses in a realistic clinical setting which can then be tested in the laboratory in appropriate models in a reverse translational approach. However, clinical trial design has had to be rethought to obtain appropriate clinical samples for laboratory studies. Presurgical or tissue-based phase Ia or IIa neoadjuvant trials can allow for evaluation of clinical signals in new tumor types, biomarker analysis and mechanistic insights, such as through immune monitoring and gene expression profiling in pre- and post-treatment tumor samples. The first of these neoadjuvant trials was in 2006, prior to any US Food and Drug Administration approvals, and assessed anti-cytotoxic T-lymphocyte-associated antigen (CTLA)-4 in 12 patients with localized bladder cancer [5]. This showed a clinical signal for safety and efficacy, with three patients achieving a pathological complete response (pCR). Increased expression of inducible costimulator (ICOS), a T-cell-specific molecule that belongs to the CD28/CTLA-4/B7 immunoglobulin superfamily, was observed in tumors treated with anti-CTLA-4 antibody. In another neoadjuvant trial in 24 patients with localized urothelial cancer, durvalumab, an anti-programmed death-ligand (PD-L)1 antibody, combined with anti-CTLA-4, resulted in a 37.5% pCR rate [6]. Increased density of tertiary lymphoid structures and increased expression of ICOS + CD4 T cells was associated with improved response. In a murine melanoma model, antitumor T-cell responses to anti-CTLA-4 were significantly reduced in ICOS-deficient versus ICOS-sufficient animals [7]. These data suggest the ICOS/ICOS ligand pathway is necessary for optimal antitumor response to anti-CTLA-4, meaning it may be a therapeutic target.

In general, higher tumor mutational burden (TMB) is associated with better response to immune checkpoint blockade. Prostate cancer is typically poorly immunogenic. In patients with metastatic castration-resistant prostate cancer, high intratumoral CD8 T cell density and an interferon (IFN)-γ response gene signature and/or antigen-specific T cell responses were associated with a favorable response to ipilimumab [8]. Mutations in prostate cancer were recognized by T cells, although the extent to which T cells infiltrate into prostate tumors is unclear. In a neoadjuvant trial of ipilimumab plus androgen deprivation therapy in patients with localized prostate cancer, an increase in CD4 and CD8 T cells, including PD-1 + and ICOS + subsets, were observed after ipilimumab [9]. There were significantly higher frequencies of CD4, CD8, and ICOS + T cells in post-treatment tumors and significant increases in tumor-infiltrating immune cells, including CD4 + , CD8 + , ICOS + , CD45RO + , granzyme-B + , and CD68 + cells. Ipilimumab resulted in increased PD-L1 and V-domain Ig suppressor of T cell activation (VISTA) expression in post-treatment prostate tumors, both of which are potent inhibitors of human T cell responses. PD-L1 + and VISTA + macrophages (CD68 +) manifest an M2-phenotype and suppress T cell function. These data suggest that an increase in immune cell infiltration may be insufficient to generate antitumor responses and blockade of other immune checkpoints such as PD-L1 may be needed to provide clinical benefit for patients with prostate cancer.

Based on these data, the combination of nivolumab 1 mg/kg plus ipilimumab 3 mg/kg were evaluated in the CheckMate 650 trial in patients with metastatic castration-resistant prostate cancer [10]. Objective response rates (ORR) were 25% in a pre-chemotherapy cohort and 10% in a post-chemotherapy cohort; four patients, two in each cohort, had complete responses. However, increased toxicity with the combination has resulted in the optimal dosing regimen being further considered.

There are multiple immune checkpoints, which are dynamic in their expression and so should be evaluated in both pre- and on-treatment tumor samples to guide clinical-decision making. Neoadjuvant trials provide clinical benefit for patients with localized disease, as well as offering mechanistic insights that can be used to develop combination therapies.

Breast cancer

Immunotherapy with PD-1/PD-L1 blockade was first approved for patients with metastatic triple-negative breast cancer (TNBC) based on the IMpassion 130 trial of atezolizumab and KEYNOTE-355 trial of pembrolizumab [11, 12]. These findings raised the questions of whether the addition of immunotherapy to chemotherapy would be more beneficial at an earlier disease stage, whether a potential improved pCR rate is correlated with improved survival, and whether or not immunotherapy before surgery is safe.

Several neoadjuvant trials in breast cancer have explored these questions. I-SPY2 was an open-label, multicenter phase II trial in which patients with high-risk stage II/III breast cancer were adaptively randomized to neoadjuvant chemotherapy with or without pembrolizumab [13]. The addition of pembrolizumab improved pCR rates versus chemotherapy in ERBB2-negative, hormone receptor-positive/ERBB2-negative, and triple-negative cohorts. In the larger phase 3 KEYNOTE-522 trial, 1174 patients with previously untreated stage II-III TNBC were randomized to neoadjuvant chemotherapy (paclitaxel and carboplatin followed by doxorubicin or epirubicin with cyclophosphamide) with either pembrolizumab or placebo [14]. After surgery, patients received adjuvant pembrolizumab or placebo for up to 27 weeks. At the first interim analysis, the pCR rate was 13.6% higher in the pembrolizumab arm (65% vs 51%; p < 0.001), with greater benefit seen in patients with clinically node-positive disease. Patients also benefited from the addition of pembrolizumab irrespective of whether their disease was PD-L1 positive or negative, which contrasts with data in the metastatic setting where benefit was limited to PD-L1 positive patients. After a median follow-up of 15.5 months, event-free survival (EFS) was higher in the pembrolizumab arm but did not reach the prespecified p-value boundary for significance. In subsequent analysis, 3-year EFS rate was significantly improved with pembrolizumab versus placebo (84.5% vs 76.8% p = 0.0003) [15]. Patients with a pCR had good survival outcomes irrespective of whether they received pembrolizumab, whereas patients without pCR had better outcomes with pembrolizumab than placebo, suggesting an additional benefit of immunotherapy in these patients. Treatment-related adverse events of grade ≥ 3 were generally similar with and without pembrolizumab in the neoadjuvant phase, although treatment discontinuations were higher. The most frequent immune-related adverse events, 10–15% of which can be permanent, were thyroid toxicities. On the basis of this trial, pembrolizumab was approved in the US for neoadjuvant use in this patient population.

In the phase III IMpassion 031 trial, 333 patients with previously untreated stage II-III TNBC were randomized to neoadjuvant chemotherapy plus atezolizumab or placebo [16]. The addition of atezolizumab to chemotherapy resulted in a significant improvement in pathological response, with pCR being achieved by 58% of patients in the atezolizumab arm versus 41% in the placebo arm (p = 0.0044). As in KEYNOTE-522, most benefit was observed in patients with node-positive disease and benefit was seen in patients regardless of PD-L1 status.

In contrast to these two studies, pCR rate was not significantly improved by the addition of neoadjuvant atezolizumab to carboplatin and nab-paclitaxel in the NeoTrip trial of 280 patients with TNBC (43.5% vs 40.8% with placebo) [17]. Unlike KEYNOTE-522 or Impassion 031, patients only received anthracycline-based therapy post-surgery, with a neoadjuvant regimen of carboplatin and nab-paclitaxel. These data called into question the optimal chemotherapy backbone to pair with immunotherapy in the preoperative setting.

In the phase II GeparNuevo trial, 174 patients with TNBC were randomized to durvalumab or placebo for 2 weeks before the addition of nab-paclitaxel followed by epirubicin plus cyclophosphamide [18]. The trial was subsequently amended so that the initial durvalumab monotherapy ‘window’ phase was removed. Overall, the pCR rate with durvalumab was 53.4% versus 44.2% with placebo, which did not meet the prespecified p-value for significance. However, a significant benefit was seen with durvalumab in the window cohort (n = 117), with a pCR of 61.0% versus 41.4%, with placebo (p = 0.035). After a median follow-up of 42.2 months, 3-year invasive disease-free survival (DFS) was 84.9% with durvalumab vs 76.9% with placebo (hazard ratio [HR] 0.54, p = 0.0559); this was achieved event though durvalumab was only administered as neoadjuvant and not adjuvant therapy [19].

Overall, the KEYNOTE-522 and IMPassion 031 studies provide consistent and complementary results that are supportive of the role of neoadjuvant immunotherapy, with both studies showing an increase in pCR and benefit observed regardless of PD-L1 status. The role of adjuvant immunotherapy in patients with a pCR remains unclear. The optimal chemotherapy regimen is also still be identified. Optimal patient selection is also important; 35–50% of patients will obtain pCR with chemotherapy alone so there is a need to identify which patients might be able to avoid immunotherapy and its associated toxicities. Biomarkers of response and toxicity are also required for those patients who might benefit from immunotherapy (Fig. 1).

Fig. 1
figure 1

Summary Neoadjuvant Immunotherapy in Breast Cancer Studies

Colon cancer

Immune checkpoint inhibition as monotherapy or in combination in metastatic colorectal cancer (CRC) has been associated with response rates of 30–70% in microsatellite-instability-high (MSI-H) or mismatch-repair-deficient (dMMR) tumors. However, response rates in mismatch-repair-proficient tumors (pMMR) are low. MMR deficient tumors account for only around 4% of all metastatic CRC, and 15% of early-stage CRC, although this still accounts for large numbers of patients.

Neoadjuvant immunotherapy may have higher efficacy in early-stage colon cancers. In the exploratory NICHE study, chemotherapy-naïve patients with dMMR or pMMR tumors received a single dose of ipilimumab and two doses of nivolumab before surgery [20]. In preliminary analyses of 40 patients, treatment was well-tolerated and all patients underwent surgery without delays. Over 80% of dMMR patients had stage III disease. Pathological response was observed in all 20 dMMR tumors, with 12 pCRs and 19 major pathological responses (MPRs), i.e., residual viable tumor of ≤ 10%. In pMMR tumors, 4 of 15 patients (27%) had a pathological response, with one partial response and three MPRs. Eleven patients were classed as non-responders, although four of these had tumor regression of over 10%. Of interest, these patients had a similar immune activation of the tumor microenvironment (TME) as dMMR responders. CD8 + PD-1 + T cell infiltration was found to be predictive of response in pMMR tumors.

Underestimation of pathological response using computed tomography (CT) scans was observed in several patients in the NICHE trial. Inflammation, acellular mucin and fibrosis accounted for the discordance between radiologic and pathologic responses. Potentially, the use of circulating tumor DNA and/or endoscopy may help in identifying patients who could avoid surgery in an organ-sparing approach.

The preliminary data from the NICHE study compare favorably with standard of care neoadjuvant chemotherapy in dMMR/MSI colon cancers. In the FOxTROT trial of 1053 patients with CRC, 95% of dMMR/MSI tumors had no response to chemotherapy [21]. However, immunotherapeutic responses need to be improved for pMMR patients, potentially through the use of biomarkers such as CD8 + /PD1 + T cells to select patients.

Non-small-cell lung cancer

MPR is predictive of long-term survival benefit in neoadjuvant chemotherapy-treated patients with NSCLC [22,23,24,25] and has been implemented as candidate surrogate endpoint for evaluating novel chemotherapies and immunotherapy response, given the rare frequency of pCR following neoadjuvant chemotherapy in NSCLC (median 4%) [2].

The rationale for neoadjuvant immune checkpoint inhibition is provided by several lines of evidence. Preclinical data demonstrated that tumor PD-L1 expression facilitates the development and spread of metastasis in murine models of NSCLC [26]. In these models, genetic and pharmacologic targeting of PD-L1 on cancer cells suppresses metastasis and PD-L1 inhibition reverses the CD8 + T cell dysfunction. These results are relevant in supporting the administration of immunotherapy in the early-stage setting, given the propensity of early NSCLC to relapse with distant metastases. In early disease, an intact immune system and minimal clonal resistance also suggest an ideal setting for optimal response to immunotherapy and to reduce risk of disease recurrence.

Several trials have indicated that neoadjuvant immunotherapy is safe and effective in patients with NSCLC. Initial studies showed that neoadjuvant nivolumab was well tolerated and induced a MPR in 45% of resected tumors [27]. Neoadjuvant atezolizumab in the LCMC3 study induced MPR in 21% of patients with a pCR rate of 7% [28]. In the phase II NEOSTAR trial, 44 patients with operable NSCLC were randomized to neoadjuvant nivolumab monotherapy or nivolumab in combination with ipilimumab with MPR rate as primary endpoint [29]. The combination arm met the trial prespecified MPR efficacy boundary to be considered promising for further testing, with a 38% MPR rate versus 22% in the nivolumab arm. Combined PD-1/CTLA-4 blockade also enhanced tumor immune infiltration and immunological memory, with increased effector, tissue-resident memory and effector memory T cells in resected tumors compared with PD-1 monotherapy. Pretreatment tumor PD-L1 was associated with response to neoadjuvant immune checkpoint inhibition and increased abundance of gut Ruminococcus and Akkermansia spp. was associated with MPR to combination therapy and with favourable T cell receptor clonality at surgery [29]. Interestingly, patients treated with neoadjuvant immunotherapy demonstrated the apparent radiological progression of the disease in lymph nodes post-therapy; however, upon pathological evaluation these lymph nodes had de novo non-caseating granulomas and were devoid of cancer; this phenomenon has been described as ‘nodal immune flare’ (NIF) [30]. NIF was associated with an inflamed nodal immune microenvironment with enrichment of antigen-presenting cells, cytotoxic and T helper 1 cell markers and immune response pathways, and with fecal abundance of Coriobacteriaceae [30]. The NIF phenomenon should be carefully distinguished from true nodal disease progression to ensure appropriate curative-intent surgical approach.

Other studies have reported that immune checkpoint blockade combined with chemotherapy or radiation has clinical activity in patients with resectable NSCLC. The addition of neoadjuvant nivolumab, atezolizumab or durvalumab to neoadjuvant chemotherapy have all shown clinical promise in small-scale phase II studies, with encouraging MPR and pCR rates and overall good tolerability [31,32,33]. Neoadjuvant durvalumab combined with stereotactic body radiotherapy induced greater MPR rates compared with durvalumab monotherapy and was well tolerated [34].

Importantly, across all these studies, neoadjuvant immunotherapy did not negatively impact surgery. Delays to surgery were infrequent and resectability rates ranged from 81 to 96%, confirming that planned surgery after neoadjuvant immunotherapy is overall feasible.

In the first phase III trial of neoadjuvant immunotherapy to report, the CheckMate-816 trial, patients with resectable NSCLC were randomized to chemotherapy with or without nivolumab [35]. In the intention-to-treat population, the addition of nivolumab to platinum-based hemotherapy increased the pCR rate, assessed in both resected primary lung tumor and sampled lymph nodes, to 24% compared with 2.2% in the chemotherapy arm (p < 0.0001). The MPR rate was 37% with the nivolumab combination versus 9% with chemotherapy alone. The pCR rate was higher with neoadjuvant nivolumab plus chemotherapy as compared with chemotherapy alone across key subgroups, including disease stage, histology, tumor PD-L1 expression and TMB [35]. Definitive surgery occurred in 83% of patients treated with nivolumab plus chemotherapy and in 75% of patients treated with chemotherapy alone [36].

In conclusion, neoadjuvant immunotherapy is safe, feasible and active in patients with resectable NSCLC. The combination of nivolumab and ipilimumab induces higher MPR rates than historical controls of neoadjuvant chemotherapy and, in an exploratory analysis, than nivolumab monotherapy, and is also associated with greater tumor immune infiltrate and immunological memory than PD-1 monotherapy. CheckMate-816 is the first phase III trial to show a pCR benefit with neoadjuvant nivolumab plus chemotherapy in resectable NSCLC, with no major impact on surgery feasibility. Preclinical and translational studies are needed to identify biomarkers of response and mechanisms of resistance to these therapies, in order to develop more effective combinatorial strategies for patients with resectable NSCLC.

Melanoma

In patients with clinical stage III melanoma, survival outcomes have historically been disappointing. Although adjuvant studies of immune- and targeted therapy have shown improved outcomes, it should be noted that these studies have screening failure rates of 15–25% due to rapid progression of disease after surgery in high-risk stage III patients. It can be postulated that patients with an aggressive disease phenotype may benefit more from a neoadjuvant approach.

In an International Neoadjuvant Melanoma Consortium (INMC) pooled analysis of six immunotherapy or BRAF targeted therapy clinical trials, pCR correlated with improved recurrence-free survival (RFS) (2-year RFS 89% vs. 50% in patients with or without pCR, p < 0.001) and overall survival (OS) (95% vs. 83%, p = 0.027) [4]. In patients with pCR, near pCR or partial pathological response with immunotherapy, very few relapses were seen, with a 2-year RFS of 96%. However, only a complete response to targeted therapy was associated with RFS, any pathological response (including a partial response) was associated with improved RFS in patients receiving neoadjuvant immunotherapy.

Several studies have investigated the effects of neoadjuvant checkpoint inhibition in melanoma. In a study of 27 patients receiving single-dose neoadjuvant plus adjuvant pembrolizumab, eight had a pCR or MPR [37]. All eight patients remained disease-free at a median follow-up of 18 months. Baseline proliferation of CD8 T cells was associated with clinical benefit and transcriptional analysis revealed a pretreatment immune signature predictive of response. In a randomized phase II study of neoadjuvant nivolumab versus ipilimumab combined with nivolumab in 23 patients with high-risk resectable melanoma, responses in the monotherapy arm were modest (ORR 25%, pCR 25%) [38]. Two patients progressed to metastatic disease during treatment and one shortly after surgery. Two patients had local recurrence after adjuvant therapy. However, the regimen was well tolerated with only 8% of patients with grade 3–4 treatment-related adverse events. In comparisons, the combination arm had high response rates (ORR 73%, pCR 45%). However, the regimen was associated with significant toxicity, with 73% of patients having grade 3–4 toxicities. Responders tended to have a higher TMB, higher PD-L1 expression and higher CD8 T cells; higher lymphoid infiltrates in responders to both therapies and a more clonal and diverse T cell infiltrate in responders to nivolumab monotherapy were also observed.

The open-label OpACIN-neo trial was designed to identify an effective but more tolerable dosing schedule of ipilimumab plus nivolumab, with patients randomized to one of three different ipilimumab plus nivolumab dosing regimens [39]. Of these, two cycles of ipilimumab 1 mg/kg plus nivolumab 3 mg/kg every 3 weeks was identified as tolerable and effective, inducing a pathological response in 77% of patients. At 2-year follow-up, almost all high-grade adverse events had resolved to ≤ grade 1, except for grade 2 endocrinopathies. More than 80% of patients were relapse-free at 2-years without adjuvant treatment, and RFS remained significantly higher for patients with pathologic response versus non-responders (97% versus 36%). High baseline TMB and IFN-γ signature expression were associated with response outcomes [40]. This is being further investigated in the DONIMI study, which will assess nivolumab alone, with a class 1 histone deacetylase inhibitor, domatinostat, or with both domatinostat and ipilimumab, in 40 patients with newly diagnosed or recurrent disease [41]. In patients with IFN-γ signature-high melanoma, radiological and pathological response rates were robust with nivolumab alone or combined with domatinostat, whereas in IFN-γ signature-low patients, response rates were low with nivolumab plus domatinostat and with nivolumab plus domatinostat plus ipilimumab.

In another recent study, 30 patients with clinical stage III or oligometastatic stage IV melanoma with Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 disease were treated with neoadjuvant followed by adjuvant nivolumab plus the anti-LAG3 antibody relatlimab [42]. The pCR rate was 59% and MPR rate was 66%. One-year RFS was 100% for patients with MPR compared to 80% for non-MPR patients. Nivolumab plus relatlimab was well tolerated, with no treatment-related grade ≥ 3 or higher toxicities noted in the neoadjuvant setting and no surgical delays due to treatment-related toxicity. In the adjuvant setting, 26% of patients had grade 3 toxicity, with adrenal insufficiency the most frequent. Increased effector CD8 + T cells and decreased immunosuppressive M2 macrophages correlated with a favorable response.

Overall, the ipilimumab 1 mg/kg plus nivolumab 3 mg/kg appear to be the most robust regimen, although nivolumab plus relatlimab appears to be comparable. Ongoing questions include whether adjuvant therapy is required after neoadjuvant treatment, especially in patients with robust pathological responses. We also need to know if neoadjuvant therapy can reduce the extent of surgery and whether baseline tumor features can be used to help to delineate which neoadjuvant treatment regimen might be most appropriate (Fig. 2).

Fig. 2
figure 2

Overview of Neoadjuvant Immunotherapy Trials in Melanoma

Non-melanoma skin cancer

Similar to melanoma, non-melanoma skin cancer (NMSC) including cutaneous squamous cell carcinoma (CSCC) has a high mutational burden, providing a strong rationale for the use of immunotherapy. Several trials have shown the efficacy of PD-1 inhibition in CSCC [43,44,45,46]. However, earlier application may potentially improve outcomes still further.

In an MD Anderson Cancer Center trial, 20 patients with primary or recurrent stage III-IVA CSCC of the head and neck (measurable disease of > 1.5 cm per RECIST 1.1) with planned curative-intent surgery and adjuvant radiation received two cycles of neoadjuvant PD-1 inhibition with cemiplimab, [47]. The primary endpoint was ORR per RECIST 1.1. Secondary endpoints included pCR, MPR, disease-specific survival (DSS), disease-free survival (DFS), and OS. Safety was assessed and biomarkers of response were evaluated.

Although only six patients (30%) had partial imaging responses by RECIST, 15 patients (75%) had either a pCR (11, 55%) or a MPR (4, 20%). This illustrates the underestimation of pathological response by radiographic imaging. Of the 15 pathologic responders, 12 (80%) did not receive adjuvant radiation as planned and at a median follow up of 22.6 months all remain free of disease. An additional 2 (10%) patients had a pPR, were treated with adjuvant therapy and also remain disease free. Three (15%) patients had > 50% viable tumor remaining at the time of surgery and were classified as non-responders. These patients had neither a clinical nor pathologic response, and all recurred. In retrospect, 2 of these patients had borderline resectable disease at presentation, underscoring the importance of careful patient selection. Histopathologic assessments showed significant heterogeneity, even among responders, including in the extent of inflammatory infiltrate. The 12-month DSS, DFS, and overall survival (OS) rates were 95%, 90%, and 95%, respectively.

Pre-treatment gene expression profiling revealed an inflamed TME in patients with pCR or MPR, indicating that a favorable immune microenvironment prior to cemiplimab treatment is associated with pathologic response. Mass cytometry CyTOF analysis of pre- and post-treatment tumor samples showed changes in the tumor immune microenvironment after cemiplimab treatment were also associated with pathologic responses, in particular enrichment of memory CD8 + T-cell clusters. A regulatory T cell subset (CD3 + CD4 + FOXP3 +) and a myeloid cell subset expressing inhibitory marker VISTA (CD68 + CD14 + VISTA +) were associated with non-response and may help explain why some patients failed treatment. A confirmatory multicenter phase II trial with 76 patients receiving neoadjuvant cemiplimab followed by surgery and adjuvant cemiplimab, radiation or observation was recently completed (clinicaltrials.gov: NCT04154943).

In conclusion, NMSC, in particular CSCC, is highly responsive to immunotherapy and the earlier application of immunotherapy may improve outcomes. Neoadjuvant immunotherapy is well tolerated and demonstrates durable responses in CSCC with no surgical delays and diminished need for adjuvant radiation therapy. Careful patient selection for neoadjuvant immunotherapy remains paramount, and a favorable baseline TME may predict response (Fig. 3).

Fig. 3
figure 3

Immunotherapy CSCC-Pathology Results. Three patients who recurred had neither clinical or pathologic response. Two patients who recurred had borderline resectable disease underscoring the importance of patient selection

Refining pathological response assessment in neoadjuvant therapy for melanoma

Pathological assessment is critical for evaluating neoadjuvant therapy and is typically the primary endpoint in clinical trials. It is therefore important that it is performed in a standardized manner; the INMC has developed guidelines for pathological evaluation of specimens to facilitate this. Tumor size assessed by radiology correlates with the tumor bed size but not the extent of residual viable tumor [48].

Reproducibility of pathological response is very important. In the OpACIN-neo trial, patients were recruited in Australia and Europe (the Netherlands and Sweden). Pathological responses were evaluated at the respective centers at which patients were recruited. Lymph node dissection specimens of 83 patients were shared across centers and re-evaluated by pathologists at the other sites, with excellent interobserver reproducibility for pathological response category and percentage pathological response being observed [48]. Standardization of pathological assessment has also enabled data to be pooled from different melanoma neoadjuvant clinical trials in order to compare efficacy of treatments [4].

Pathological response is an excellent predictor of clinical outcome in melanoma patients treated with neoadjuvant therapies. As a consequence, pathological response evaluation enables clinicians to provide information to patients on their long-term prognosis after a short period of neoadjuvant therapy and may also inform clinical decision-making. In the PRADO trial of combination neoadjuvant nivolumab and ipilimumab, patients with a pCR or near pCR in the index node were followed-up with observation only, whereas patients with a partial or no response underwent therapeutic lymph node dissection (TLND); patients with no response in their TLND then received adjuvant nivolumab or dabrafenib plus trametinib while partial response patients underwent observation [49]. Thus, pathological response is being used to personalize treatment and de-escalate surgery. At a minimum of 12 weeks follow-up, 70 of 99 patients (71%) had a pathological response in the index lymph node and 60 patients (61%) had MPR in the index lymph node and did not undergo therapeutic lymph node dissection. These patients had fewer surgery-related adverse events and much higher quality-of-life scores.

In OpACIN-neo, almost all patients with recurrence had ≥ 70% residual viable melanoma. Type of pathological response also predicted outcome, with patients with a high fibrosis subtype of treatment response having the strongest association with lack of recurrence and prolonged RFS. These data are being used to develop a predictive score for relapse [48]. Patients with a pCR or near pCR had a high degree of hyaline fibrosis. However, pigment-laden macrophages, necrosis and tumor-infiltrating T cell infiltrate did not correlate with category of pathological response. Even in patients with no pathological response, hyaline fibrosis is associated with prolonged RFS. Similarly, patients treated with neoadjuvant dabrafenib and trametinib who achieved pCR had longer RFS. Hyalinized fibrosis was again associated with longer RFS in patients whereas necrosis and/or immature/proliferative fibrosis correlated with shorter RFS [50]. Patients with high fibrosis have differences in circulating T, natural killer (NK) and myeloid cells in peripheral blood compared with low fibrosis patients. Subtype of pathological response following neoadjuvant therapy appears to be important in determining outcomes. High TMB and high IFN-γ signature score are also associated with pathologic response and low risk of relapse [48]. In addition, gastrointestinal microbial diversity is low in both non-responders and patients with severe immune-related adverse events.

In conclusion, pathological response assessment is reproducible. Radiology tumor size correlates with the tumor bed size but not pathological response. Pathological, peripheral blood, molecular and microbiome-related factors may all be associated with response and prolonged survival.

Conclusions

Administration of immunotherapy in a neoadjuvant setting offers the potential to help improve outcomes for patients with high-risk, surgically resectable cancers. Neoadjuvant immunotherapy may improve operability, prime the antitumor immune response at an early stage, minimise micrometastatic disease, and prevent tumor relapse. Additionally, neoadjuvant treatment also provides an opportunity for early assessment of tumor response to therapy using surrogate endpoints of clinical efficacy at surgery. Neoadjuvant immunotherapy rather than upfront surgery and adjuvant therapy has the potential to become a new standard of care in several settings.

The Neoadjuvant Immunotherapy Across Cancers at the 2021 Immunotherapy Bridges meeting provided an invaluable opportunity to bring together experts in immunotherapy and neoadjuvant treatment. Highlighted in this session were changes to standard of care as well as ongoing trials which continue to evaluate neoadjuvant immunotherapies in different tumors. This session also highlighted the neoadjuvant platform as a unique, pan-cancer opportunity to leverage biomarker analyses from robust tissue collections to gain novel insights into mechanisms of response and resistance to therapies, potentially informing subsequent development of immunotherapy in other indications. While surrogate endpoint validation for chemotherapy is well established, pathologic response to neoadjuvant immunotherapy is in development and will be critical to placing biomarker insights, clinical response, and outcomes into context. Treatment of surgically resectable cancers with neoadjuvant immunotherapy remains of significant interest to improve outcomes for patients across cancer types and stages.

Availability of data and materials

Not applicable.

Abbreviations

CRC:

Colorectal cancer

CSCC:

Cutaneous squamous cell carcinoma

CT:

Computed tomography

CTLA-4:

Cytotoxic T-lymphocyte-associated antigen-4

dMMR:

Mismatch-repair-deficient

DFS:

Disease-free survival

DSS:

Disease-specific survival

EFS:

Event-free survival

HR:

Hazard ratio

ICOS:

Inducible T-cell costimulator

IFN:

Interferon

INMC:

International Neoadjuvant Melanoma Consortium

MPR:

Major pathological response

MSI:

Microsatellite-instability-high

NIF:

Nodal immune flare

NK:

Natural killer

NMSC:

N-melanoma skin cancer

NSCLC:

Non-small-cell lung cancer

ORR:

Objective response rate

OS:

Overall survival

pMMR:

Mismatch-repair-proficient

pCR:

Pathological complete response

PD:

Programmed death

PD-L1:

Programmed death ligand

RECIST:

Response evaluation criteria in solid tumors

RFS:

Recurrence-free survival

TLND:

Therapeutic lymph node dissection

TMB:

Tumor mutational burden

TME:

Tumor microenvironment

TNBC:

Triple-negative breast cancer

VISTA:

V-domain Ig suppressor of T cell activation

References

  1. Liu J, Blake SJ, Yong MC, Harjunpää H, Ngiow SF, Takeda K, et al. Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discov. 2016;6(12):1382–99.

    Article  CAS  PubMed  Google Scholar 

  2. Hellmann MD, Chaft JE, William WN Jr, Rusch V, Pisters KM, Kalhor N, et al. Pathological response after neoadjuvant chemotherapy in resectable non-small-cell lung cancers: proposal for the use of major pathological response as a surrogate endpoint. Lancet Oncol. 2014;15(1):e42-50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Huang M, O’Shaughnessy J, Zhao J, Haiderali A, Cortes J, Ramsey S, et al. Evaluation of pathologic complete response as a surrogate for long-term survival outcomes in triple-negative breast cancer. J Natl Compr Canc Netw. 2020;18(8):1096–104.

    Article  PubMed  Google Scholar 

  4. Menzies AM, Amaria RN, Rozeman EA, Huang AC, Tetzlaff MT, van de Wiel BA, et al. Pathological response and survival with neoadjuvant therapy in melanoma: a pooled analysis from the International Neoadjuvant Melanoma Consortium (INMC). Nat Med. 2021;27(2):301–9.

    Article  CAS  PubMed  Google Scholar 

  5. Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso P, et al. CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci USA. 2008;105(39):14987–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gao J, Navai N, Alhalabi O, Siefker-Radtke A, Campbell MT, Tidwell RS, et al. Neoadjuvant PD-L1 plus CTLA-4 blockade in patients with cisplatin-ineligible operable high-risk urothelial carcinoma. Nat Med. 2020;26(12):1845–51.

    Article  CAS  PubMed  Google Scholar 

  7. Fu T, He Q, Sharma P. The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res. 2011;71(16):5445–54.

    Article  CAS  PubMed  Google Scholar 

  8. Subudhi SK, Vence L, Zhao H, Blando J, Yadav SS, Xiong Q, et al. Neoantigen responses, immune correlates, and favorable outcomes after ipilimumab treatment of patients with prostate cancer. Sci Transl Med. 2020;12(537):eaaz3577.

    Article  CAS  PubMed  Google Scholar 

  9. Gao J, Ward JF, Pettaway CA, Shi LZ, Subudhi SK, Vence LM, et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med. 2017;23(5):551–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sharma P, Pachynski RK, Narayan V, Fléchon A, Gravis G, Galsky MD. Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 trial. Cancer Cell. 2020;38(4):489–99.

    Article  CAS  PubMed  Google Scholar 

  11. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, et al. Atezolizumab and Nab-Paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379(22):2108–21.

    Article  CAS  PubMed  Google Scholar 

  12. Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im SA, Yusof MM, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396(10265):1817–28.

    Article  PubMed  Google Scholar 

  13. Nanda R, Liu MC, Yau C, Shatsky R, Pusztai L, Wallace A, et al. Effect of pembrolizumab plus neoadjuvant chemotherapy on pathologic complete response in women with early-stage breast cancer: an analysis of the ongoing phase 2 adaptively randomized I-SPY2 trial. JAMA Oncol. 2020;6(5):676–84.

    Article  PubMed  Google Scholar 

  14. Schmid P, Cortes J, Pusztai L, McArthur H, Kümmel S, Bergh J, et al. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382(9):810–21.

    Article  CAS  PubMed  Google Scholar 

  15. Schmid P, Cortes J, Dent R, Pusztai L, McArthur H, Kümmel S, et al. Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med. 2022;386(6):556–67.

    Article  CAS  PubMed  Google Scholar 

  16. Mittendorf EA, Zhang H, Barrios CH, Saji S, Jung KH, Hegg R, et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet. 2020;396(10257):1090–100.

    Article  CAS  PubMed  Google Scholar 

  17. Gianni L, Huang C-S, Egle D, Bermejo B, Zamagni C, Thill M, Anton A, et al. Pathologic complete response (pCR) to neoadjuvant treatment with or without atezolizumab in triple negative, early high-risk and locally advanced breast cancer .NeoTRIPaPDL1 Michelangelo randomized study. Cancer Res. 2020. https://doi.org/10.1158/1538-7445.SABCS19-GS3-04.

    Article  Google Scholar 

  18. Loibl S, Untch M, Burchardi N, Huober J, Sinn BV, Blohmer JU, et al. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: clinical results and biomarker analysis of GeparNuevo study. Ann Oncol. 2019;30(8):1279–88.

    Article  CAS  PubMed  Google Scholar 

  19. Loibl S, Schneeweiss A, Huober JN, Braun M, Rey J, Blohmer JU, et al. Durvalumab improves long-term outcome in TNBC: results from the phase II randomized GeparNUEVO study investigating neodjuvant durvalumab in addition to an anthracycline/taxane based neoadjuvant chemotherapy in early triple-negative breast cancer (TNBC). J Clin Oncol. 2021;39(15):506–506.

    Article  Google Scholar 

  20. Chalabi M, Fanchi LF, Dijkstra KK, Van den Berg JG, Aalbers AG, Sikorska K, et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med. 2020;26(4):566–76.

    Article  CAS  PubMed  Google Scholar 

  21. Morton D. FOxTROT: an international randomised controlled trial in 1053 patients evaluating neoadjuvant chemotherapy (NAC) for colon cancer. On behalf of the FOxTROT collaborative group. Ann Oncol. 2019;30(5):v198.

    Article  Google Scholar 

  22. Chaft JE, Rusch V, Ginsberg MS, Paik PK, Finley DJ, Kris MG, et al. Phase II trial of neoadjuvant bevacizumab plus chemotherapy and adjuvant bevacizumab in patients with resectable nonsquamous non-small-cell lung cancers. J Thorac Oncol. 2013;8(8):1084–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cascone T, Gold KA, Swisher SG, Liu DD, Fossella FV, Sepesi B, et al. Induction cisplatin docetaxel followed by surgery and erlotinib in non-small cell lung cancer. Ann Thorac Surg. 2018;105(2):418–24.

    Article  PubMed  Google Scholar 

  24. Cascone T, Sepesi B, Lin HY, Kalhor N, Parra ER, Jiang M, et al. A phase I/II study of neoadjuvant cisplatin, docetaxel, and nintedanib for resectable non-small cell lung cancer. Clin Cancer Res. 2020;26(14):3525–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Pataer A, Weissferdt A, Vaporciyan AA, Correa AM, Sepesi B, Wistuba II, et al. Evaluation of pathologic response in lymph nodes of patients with lung cancer receiving neoadjuvant chemotherapy. J Thorac Oncol. 2021;16(8):1289–97.

    Article  PubMed  Google Scholar 

  26. Chen L, Gibbons DL, Goswami S, Cortez MA, Ahn YH, Byers LA, et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun. 2014;5:5241.

    Article  CAS  PubMed  Google Scholar 

  27. Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. 2018;378(21):1976–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lee J, Chaft J, Nicholas A, Patterson A, Waqar S, Toloza E, et al. Surgical and clinical outcomes with neoadjuvant atezolizumab in resectable Stage IB–IIIB NSCLC: LCMC3 trial primary analysis. J Thor Oncol. 2021;16(3 Suppl):S59–61.

    Article  Google Scholar 

  29. Cascone T, William WN Jr, Weissferdt A, Leung CH, Lin HY, Pataer A, et al. Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: the phase 2 randomized NEOSTAR trial. Nat Med. 2021;27(3):504–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cascone T, Weissferdt A, Godoy MCB, William WN Jr, Leung CH, Lin HY, et al. Nodal immune flare mimics nodal disease progression following neoadjuvant immune checkpoint inhibitors in non-small cell lung cancer. Nat Commun. 2021;12(1):5045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Provencio M, Nadal E, Insa A, García-Campelo MR, Casal-Rubio J, Dómine M, et al. Neoadjuvant chemotherapy and nivolumab in resectable non-small-cell lung cancer (NADIM): an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020;21(11):1413–22.

    Article  CAS  PubMed  Google Scholar 

  32. Shu CA, Gainor JF, Awad MM, Chiuzan C, Grigg CM, Pabani A, et al. Neoadjuvant atezolizumab and chemotherapy in patients with resectable non-small-cell lung cancer: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020;21(6):786–95.

    Article  CAS  PubMed  Google Scholar 

  33. Rothschild SI, Zippelius A, Eboulet EI, Savic Prince S, Betticher D, Bettini A, et al. SAKK 16/14: durvalumab in addition to neoadjuvant chemotherapy in patients with stage IIIA(N2) non-small-cell lung cancer-a multicenter single-arm phase II trial. J Clin Oncol. 2021;39(26):2872–80.

    Article  CAS  PubMed  Google Scholar 

  34. Altorki NK, McGraw TE, Borczuk AC, Saxena A, Port JL, Stiles BM, et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol. 2021;22(6):824–35.

    Article  CAS  PubMed  Google Scholar 

  35. Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. Nivolumab (NIVO) + platinum-doublet chemotherapy (chemo) vs chemo as neoadjuvant treatment (tx) for resectable (IB-IIIA) non-small cell lung cancer (NSCLC) in the phase 3 CheckMate 816 trial. In: Proceedings of the 112th Annual Meeting of the American Association for Cancer Research; 2021 April 10–15. Philadelphia (PA): AACR; 2021. Abstract CT003.

  36. Spicer J, Wang C, Tanaka F, Saylors GB, Chen K-N, Liberman M, et al. Surgical outcomes from the phase 3 CheckMate 816 trial: Nivolumab (NIVO) + platinum-doublet chemotherapy (chemo) vs chemo alone as neoadjuvant treatment for patients with resectable non-small cell lung cancer (NSCLC). J Clin Oncol. 2021;39(15):8503–8503.

    Article  Google Scholar 

  37. Huang AC, Orlowski RJ, Xu X, Mick R, George SM, Yan PK, et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med. 2019;25(3):454–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Amaria RN, Reddy SM, Tawbi HA, Davies MA, Ross MI, Glitza IC, et al. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med. 2018;24(11):1649–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Rozeman EA, Menzies AM, van Akkooi ACJ, Adhikari C, Bierman C, van de Wiel BA, et al. Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, randomised, controlled trial. Lancet Oncol. 2019;20(7):948–60.

    Article  CAS  PubMed  Google Scholar 

  40. Rozeman EA, Hoefsmit EP, Reijers ILM, Saw RPM, Versluis JM, Krijgsman O, et al. Survival and biomarker analyses from the OpACIN-neo and OpACIN neoadjuvant immunotherapy trials in stage III melanoma. Nat Med. 2021;27(2):256–63.

    Article  CAS  PubMed  Google Scholar 

  41. Blank CU, Reijers ILM, Versluis JM, Menzies AM, Dimitriadis P, WoutersMW, et al. Personalized combination of neoadjuvant domatinostat, nivolumab (NIVO) and ipilimumab (IPI) in stage IIIB-D melanoma patients (pts) stratified according to the interferon-gamma signature (IFN-γ sign): The DONIMI study. Ann Oncol. 2021;32(5):S1283–346.

    Google Scholar 

  42. Amaria RN, Postow MA, Tetzlaff MT, Ross MI, Glitza IC, McQuade JL, et al. Neoadjuvant and adjuvant nivolumab (nivo) with anti-LAG3 antibody relatlimab (rela) for patients (pts) with resectable clinical stage III melanoma. J Clin Oncol. 2021;39(15):9502–9502.

    Article  Google Scholar 

  43. Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375(19):1856–67.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD, et al. PD-1 Blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N Engl J Med. 2018;379(4):341–51.

    Article  CAS  PubMed  Google Scholar 

  45. Migden MR, Khushalani NI, Chang ALS, Lewis KD, Schmults CD, Hernandez-Aya L, et al. Cemiplimab in locally advanced cutaneous squamous cell carcinoma: results from an open-label, phase 2, single-arm trial. Lancet Oncol. 2020;21(2):294–305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hughes BGM, Munoz-Couselo E, Mortier L, Bratland Å, Gutzmer R, Roshdy O, et al. Pembrolizumab for locally advanced and recurrent/metastatic cutaneous squamous cell carcinoma (KEYNOTE-629 study): an open-label, nonrandomized, multicenter, phase II trial. Ann Oncol. 2021;32(10):1276–85.

    Article  CAS  PubMed  Google Scholar 

  47. Ferrarotto R, Amit M, Nagarajan P, Rubin ML, Yuan Y, Bell D, et al. Pilot Phase II Trial of neoadjuvant immunotherapy in locoregionally advanced, resectable cutaneous squamous cell carcinoma of the head and neck. Clin Cancer Res. 2021;27(16):4557–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rawson RV, Adhikari C, Bierman C, Lo SN, Shklovskaya E, Rozeman EA, et al. Pathological response and tumour bed histopathological features correlate with survival following neoadjuvant immunotherapy in stage III melanoma. Ann Oncol. 2021;32(6):766–77.

    Article  CAS  PubMed  Google Scholar 

  49. Blank CH, Reijers ILM, Pennington T, et al. First safety and efficacy results of PRADO: a phase II study of personalized response-driven surgery and adjuvant therapy after neoadjuvant ipilimumab (IPI) and nivolumab (NIVO) in resectable stage III melanoma. J Clin Oncol. 2020;38(15):10002–10002.

    Article  Google Scholar 

  50. Tetzlaff MT, Adhikari C, Lo S, Rawson RV, Amaria RN, Menzies AM, et al. Histopathological features of complete pathological response predict recurrence-free survival following neoadjuvant targeted therapy for metastatic melanoma. Ann Oncol. 2020;31(11):1569–79.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

A special thanks to 3P Solution for their support and cooperation in organizing the meeting.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

EMB prepared the manuscript collaboratively with input of RNA, TC, MC, NG, EAM, RAS, PS and PAA. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Elizabeth M. Burton.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Elizabeth M. Burton (EMB): nothing to disclose. Rodabe N. Amaria (RNA): Research funding and advisory board: Novartis, BMS, Iovance. Tina Cascone (TC): speaker fees/honoraria: The Society for Immunotherapy of Cancer, Bristol Myers Squibb, Roche, Medscape, and PeerView; advisory role/consulting fees: MedImmune/AstraZeneca, Bristol Myers Squibb, EMD Serono, Merck & Co., Genentech, and Arrowhead Pharmaceuticals; Institutional research funding: MedImmune/AstraZeneca, Bristol Myers Squibb, and EMD Serono. Myriam Chalabi (MC): MSD, BMS, Roche-Genentech (research funding to the institute). Neil Gross (NG): Research Support, Consulting: Regeneron; Ad-hoc Scientific Advisory Board: Sanofi- Genzyme, Shattuck Labs; Scientific Advisory Board: PDS Biotechnology; Consulting: DragonFly, Intuitive Surgical; Institutional Consulting: Verb Surgical. Elizabeth A. Mittendorf (EAM): Scientific Advisory Board: AstraZeneca, Genomic Health, Merck, Peregrine Pharmaceuticals, Roche/Genentech, Sellas lifesciences, TapImmune; Clinical Trial Support (to institution): AstraZeneca, EMD Serono, Galena Biopharma, Roche/Genentech; Research Support: Glaxo SmithKline; Steering Committees: BMS, Lilly, Roche/Genentech Richard A. Scolyer (RAS): has received fees for professional services from F. Hoffmann-La Roche Ltd, Evaxion, Provectus Biopharmaceuticals Australia, Qbiotics, Novartis, Merck Sharp & Dohme, NeraCare, AMGEN Inc., Bristol-Myers Squibb, Myriad Genetics and GlaxoSmithKline. Funding: is supported by an NHMRC Practitioner Fellowship (APP1141295). Padmanee Sharma (PS): Consulting or Stock Ownership or Advisory Board: Achelois, Adaptive Biotechnologies, Affini-T, Apricity, BioAtla, BioNTech, Candel Therapeutics, Catalio, Codiak, Dragonfly, Earli, Enable Medicine, Glympse, Hummingbird, ImaginAb, Infinity Pharma, JSL Health, Lava Therapeutics, Lytix, Marker, Oncolytics, PBM Capital, Phenomic AI, Polaris Pharma, Sporos, Time Bioventures, Trained Therapeutix, Two Bear Capital, Venn Biosciences. Paolo A. Ascierto (PAA): Employment or Leadership Position: None; ​ Consultant/Advisory Role: Bristol-Myers Squibb, Roche-Genentech, Merck Sharp & Dohme, Novartis, Merck Serono, Pierre-Fabre, AstraZeneca, Sun Pharma, Sanofi, Idera, Sandoz, Immunocore, 4SC, Italfarmaco, Nektar, Boehringer-Ingelheim, Eisai, Regeneron, Daiichi Sankyo, Pfizer, OncoSec, Nouscom, Lunaphore, Seagen, iTeos, Medicenna; Stock Ownership: None; Research Funding: Bristol-Myers Squibb, Roche-Genentech, Pfizer, Sanofi; Expert Testimony: None; Other Remuneration: None.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Burton, E.M., Amaria, R.N., Cascone, T. et al. Neoadjuvant immunotherapy across cancers: meeting report from the Immunotherapy Bridge—December 1st–2nd, 2021. J Transl Med 20, 271 (2022). https://doi.org/10.1186/s12967-022-03472-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-022-03472-x

Keywords