Vaccine for triple-negative breast cancer produces ‘exciting’ results in early testing


Preliminary results of a phase 1 trial for a novel vaccine that could eventually prevent triple-negative breast cancer exceeded expectations, based on data presented at San Antonio Breast Cancer Symposium.

Researchers at Cleveland Clinic tested the vaccine on 16 women and 75% showed an immunologic response to the drug, which is based on the “retired protein hypothesis” developed by Vincent Tuohy, PhD, immunologist at Cleveland Clinic’s Lerner Research Institute, who died in January.

Quote from Justin M. Johnson, PhD

Study investigators also determined a safe dose of the vaccine with no adverse reactions but have reopened the trial to test new dosing levels to potentially increase its effectiveness.

“There was no guarantee we would get any type of immune response in these women, and our other fear was, what if we could only get an immune response at an unacceptably toxic dose? Then you wouldn’t be able to use it,” Justin M. Johnson, PhD, program manager at Cleveland Clinic, told Healio. “The great news is we have a dose that gives a good immune response and is well tolerated, and we’re working on fine-tuning that to be even better.”

The vaccine targets alpha-lactalbumin, a protein in both human and cow milk, that is expressed in 70% of triple-negative breast cancer tumors, according to background research provided by the researchers. Patients in this portion of the trial had been diagnosed with triple-negative breast cancer, treated within the previous 3 years and developed residual disease.

Johnson spoke with Healio about the phase 1 results, potential concerns, the future of the trial and more.

Healio: How would you summarize the results from this portion of the trial?

Johnson: The toxicity at the current dose level we plan to use is acceptable and it’s mild and well tolerated (10 µg of the alpha-lactoalbumin dose, 10 µg zymosan, which helps start the immune reaction). Second, the T-cell profile in the majority of the women tested so far has been significant. The third takeaway is that we still don’t necessarily have the optimal dose, so we’re currently testing those intermediate doses.

Healio: Why are you continuing your evaluation of the dosage?

Johnson: We thought we’d have to go much higher than 10 mg to get a response, but we are seeing a response at 10 µg. Dose level 2 was too high — the 100 µg alpha-lactalbumin proved to be a problem in only one of the six subjects tested but — per protocol — that is considered unacceptable. Now we’re looking at alpha-lactalbumin levels between 10 and 100 µg, and we’re also going to explore elevating the zymosan. We know 100 µg zymosan is too much and 10 µg works, but there might be a dose in between that’s better. For our intermediate doses, we’re going to not only raise the alpha-lactalbumin, but we may raise the zymosan as well.

Healio: How did the research team react to the results?

Johnson: I would say most of us would feel this is a better-than-expected outcome because this drug has never been tested in humans. We remained hopeful that we could get an immune response to alpha-lactalbumin in humans but, going into this, we didn’t know whether this would occur. That’s why we’re doing the trial, and the fact that roughly three-quarters of the women in our study had an immune response to the vaccine is really exciting.

Healio: Did you have concerns about irritation at the injection sites, the adverse reaction reported in the results?

Johnson: The irritation at the injection sites was expected because of the nature of this drug. It’s designed not to be immediately absorbed by the body, but rather to stay at the site of injection for several weeks, and that is intentional because that’s what really gets the immune system to notice it and to get it revved up. Hopefully, in the future, as we develop this drug even more, we’d like to be able to get rid of that side effect entirely, but for right now it’s considered a grade 1 adverse event. I’d love for the adverse events grade to go from 1 to 0; I don’t know if that’s possible. There’s nothing disappointing so far — we’re keeping our fingers crossed.

Healio: Can you describe the other cohorts in phase 1 of this trial?

Johnson: Our phase 1B trial is immunizing healthy women now that we’ve established a safe toxicity profile. These are women who have family history or genetic risk markers for breast cancer, so they have a high chance of developing triple-negative breast cancer, and they’re so concerned about it they’ve already made the decision to go to Cleveland Clinic and get a prophylactic mastectomy because of this risk. Phase 1B is designed so that we immunize the women before their surgery and then during their surgery, we take a sample of the mammary tissue. First, this makes the trial extra safe for these women because even if there were an adverse reaction in mammary tissue that we didn’t anticipate, that mammary tissue is coming out anyway. Second, it gives us a unique opportunity to look at healthy mammary tissue from women who’ve had the vaccine again to see if there’s any unusual toxicity that we didn’t anticipate.

The phase 1C portion of the trial, which we’ve also just started, came about because there was a change in the standard of care. When we designed and started this trial in 2020 and 2021, pembrolizumab (Keytruda, Merck) was not part of the routine treatment of these patients, and now it is. We have to consider what the combination of that drug and our vaccine might be. In the 1C cohort, we’re treating patients who are being treated with pembrolizumab concurrently.

Healio: Do you have any other concerns?

Johnson: We’re looking into ways to produce the drug in a more practical way. Right now, the drug is essentially made to order. It’s made in the pharmacy, and its stability is fairly short. We have to administer it within 2 hours, so we’re looking to improve that for phase 2 and beyond. To come up with a more stable formulation — one that we could put in a bottle and ship out to all of our future phase 2 testing sites and can sit on the shelf a couple of weeks and then be administered — is one of the challenges we’re dealing with right now.

I guess one disappointing thing about phase 1 is that despite the enormous interest and the number of women who want to get into the trial, accrual to phase 1B has been challenging because of the specific requirements. There’s a limited population of women who are coming in to get a prophylactic mastectomy, they must do that at Cleveland Clinic, and on top of that they have to consent to the trial. The biggest challenge is having to time these immunizations, which have to occur within a certain window before their surgery. It took us a long time to get women into that cohort after we opened it — almost a year — but I’m happy to report that we already have one woman in phase 1B and another one who will be coming on study soon. We hope to keep recruiting as many of those women as we can.

Healio: Is there anything else you’d like to say?

Johnson: I would like to mention my mentor, Dr. Tuohy, who passed away. We’re carrying on his work and his legacy. His retired protein hypothesis — which is the foundation of the work that we’re talking about — he felt was applicable to other cancer vaccines. This idea of using a protein that’s no longer normally made in the body, but is made in tumors, provides a perfect opportunity to make a potentially safe and effective vaccine. We’re developing an ovarian cancer vaccine based on this same technology, and next year we’re going to start a program where we’re looking at additional cancers — other types of cancers of the breast and ovary and also cancers in other organs.

Bioengineering Breakthrough: Tobacco Plants Synthesize Vaccine Powerhouse QS-21


A novel method for producing the key vaccine ingredient QS-21 in tobacco plants has been developed, offering a sustainable alternative to traditional extraction from the soapbark tree and enhancing vaccine manufacturing. Credit: SciTechDaily.com

Soap bark discovery offers a sustainability booster for the global vaccine market, opening unprecedented opportunities for bioengineering vaccine adjuvants.

A valuable molecule sourced from the soapbark tree and used as a key ingredient in vaccines, has been replicated in an alternative plant host for the first time, opening unprecedented opportunities for the vaccine industry.

A research collaboration led by the John Innes Centre used the recently published genome sequence of the Chilean soapbark tree (Quillaja saponaria) to track down and map the elusive genes and enzymes in the complicated sequence of steps needed to produce the molecule QS-21.

By utilizing the soapbark tree’s genome, the researchers have opened up new possibilities for bioengineering vaccine adjuvants, potentially improving vaccine efficacy and reducing environmental impact.

Advancing Vaccine Development

Using transient expression techniques developed at the John Innes Centre, the team reconstituted the chemical pathway in a tobacco plant, demonstrating for the first time ‘free-from ‘tree’ production of this highly valued compound.

Professor Anne Osbourn FRS, group leader at the John Innes Centre said: “Our study opens unprecedented opportunities for bioengineering vaccine adjuvants. We can now investigate and improve these compounds to promote the human immune response to vaccines and produce QS-21 in a way that does not depend on extraction from the soapbark tree.”

Vaccine adjuvants are immunostimulants that prime the body’s response to the vaccine – and are a key ingredient of human vaccines for shingles, malaria, and others under development.

Sustainable Solutions for Vaccine Ingredients

QS-21, a potent adjuvant, is sourced directly from the bark of the soapbark tree, raising concerns about the environmental sustainability of its supply.

For many years researchers and industrial partners have been looking for ways to produce the molecule in an alternative expression system such as yeast or tobacco plants. However, the complex structure of the molecule and lack of knowledge about its biochemical pathway in the tree have so far prevented this.

Previously researchers in the group of Professor Osbourn had assembled the early part of the pathway that makes up the scaffold structure for QS-21. However, the search for the longer full pathway, the acyl chain which forms one crucial part of the molecule that stimulates immune cells, remained unfinished.

In a new study that will be published today (January 26) in Nature Chemical Biology, researchers at the John Innes Centre used a range of gene discovery approaches to identify around 70 candidate genes and transferred them to tobacco plants.

By analyzing gene expression patterns and products, supported by the Metabolomic and Nuclear Magnetic Resonance (NMR) platforms at the John Innes Centre, they were able to narrow the search down to the final 20 genes and enzymes that make up the QS-21 pathway.

First author Dr Laetitia Martin said: “This is the first time QS-21 has been produced in a heterologous expression system. This means we can better understand how this molecule works and how we might address issues of scale and toxicity.

“What is so rewarding is that this molecule is used in vaccines and by being able to make it more sustainably my project has an impact on people’s lives. It’s amazing to think that something so scientifically rewarding can bring such good to society.”

“On a personal level, this research was scientifically extremely rewarding. I am not a chemist so I could not have done this without the support of the John Innes Centre metabolomics platform and chemistry platform.”

The team has partnered with Plant Bioscience Limited PBL (Plant Bioscience Limited) Norwich Limited who are leading the commercialization of this project.

Vaccine ‘Promising’ for Delaying Relapse in KRAS-Mutated Pancreatic Cancer, CRC


T-cell response linked to 86% reduced risk of relapse, death

A computer rendering of a T-cell killing a cancer cell.

An investigational therapeutic vaccine appeared to be effective in delaying relapse of KRAS-mutated pancreatic and colorectal cancer (CRC), according to results from the phase I AMPLIFY-201 study.

Among 25 patients with pancreatic ductal adenocarcinoma (PDAC) and CRC who were considered at high risk for relapse and who received a maximum of 10 doses of the ELI-002 vaccine targeted toward KRAS G12D and G12R mutations, T-cell responses occurred in 84%, and in 100% of those in the two highest dose cohorts, reported Shubham Pant, MD, MBBS, of the University of Texas MD Anderson Cancer Center in Houston, and colleagues.

Furthermore, T-cell response correlated with an 86% reduction in risk of relapse or death, they noted in Nature Medicineopens in a new tab or window.

“Overall, this study provides important proof of concept for the safety and immunogenicity of lymph-node-targeting Amph [Amphiphile] vaccines and yielded promising signals of clinical activity that correlates with the magnitude of ELI-02 2P-induced T cell response,” the authors wrote.

“Pancreatic cancer patients, even after resection, have a high risk of relapse even after adjuvant therapy,” Pant told MedPage Today. “So this is an area of unmet need.”

These results “are early, but promising,” he added. “If this is validated in a bigger trial, we are truly moving the needle in these patients, especially those with pancreatic cancer.”

ELI-002 is composed of AMP-modified mutant KRAS peptide antigens and ELI-004 — an AMP-modified immune-stimulatory oligonucleotide CpG adjuvant available as an off-the-shelf subcutaneous administration. Pant explained that what distinguishes ELI-oo2 as a vaccine is that it is directed into lymph nodes in order to elicit more robust immune responses and target mutant KRAS with greater potency and precision.

Of the 25 patients in the trialopens in a new tab or window (median age 61 years, 60% women), 20 had pancreatic cancer and five had CRC. Of these patients, 84% were white, 8% were Asian, and the remainder had unreported ethnicity. All 25 previously had surgery or another procedure designed to be curative, and seven previously had received radiation therapy.

The vaccine was evaluated at dose levels of 0.1, 0.5, 2.5, 5, and 10 mg. T-cell responses were observed in 21 of the 25 patients, and at the two highest dose levels, all patients (11 of 11) demonstrated elevated mutant KRAS-specific T-cell responses following ELI-002 vaccination.

Pant and colleagues observed that efficacy correlated with T-cell responses above or below the median-fold increase (12.75-fold) over baseline. Above-median T-cell response correlated with biomarker-assessed clinical antitumor activity, with a median change from baseline in tumor biomarker of -76% versus -10.2% in those patients with a below-median T-cell response (P<0.0014).

Among the 13 above-median T-cell responders, 100% showed biomarker reduction, with six achieving biomarker clearance. None of the 12 with below-median T-cell response achieved clearance, while eight showed reduction.

These findings further correlated to relapse-free survival (RFS). At at median follow-up of 8.5 months, the median RFS for the entire study cohort was 16.33 months, with the median RFS was not reached among above-median T-cell responders compared with 4.01 months in below-median T-cell responders, translating into an 86% reduced risk of relapse or death (HR 0.14, 95% CI 0.03-0.63, P=0.0167).

Regarding safety, 12 patients experienced an adverse event related to the vaccine, all grade 1 or grade 2. There were no reported cases of cytokine release syndrome and no dose-limiting toxicities. The most common adverse events of any grade were fatigue (24%), injection site reaction (16%), and myalgia (12%).

One serious adverse event was reported: a grade 3 abdominal wall hematoma that resolved 8 days after onset and was considered related to a biopsy performed per protocol to confirm progression. No events led to discontinuation of treatment or death.

Pant noted that a potential advantage of ELI-002 is its availability as an off-the-shelf therapy.

“Recently, individualized neoantigen cancer vaccines have shown promising efficacy for PDAC, non-small cell lung cancer, colorectal cancer, and melanoma,” the authors wrote. “The availability of ELI-002 as an ‘off-the-shelf’ product offers several further advantages, including streamlined standard manufacturing to facilitate on-demand availability while eliminating the need for tumor-informed production, which presents operational risks and limits use to post-surgical adjuvant stage.”

Results from this trial have led to a phase II trial opens in a new tab or windowthat will begin later this year, Pant noted, with a new formulation of ELI-002 that will target seven of the most common KRAS mutations.

A vaccine for glioma


Despite substantial advances in understanding of the molecular features of gliomas, the therapeutic options for these aggressive tumors remain scarce. Rich, Mitchell and colleagues provide their views about a phase 1 clinical trial testing the safety and efficacy of vaccines against cancer expressing mutant metabolic enzyme IDH1 in patients with high-grade glioma.

From the bench: Sameer Agnihotri and Jeremy N. Rich

Gliomas are the most prevalent primary brain tumors and remain incurable despite extensive molecular characterization and research aimed at identifying viable therapeutic vulnerabilities. Among the various glioma subtypes, diffuse gliomas and secondary glioblastomas are driven mostly by gain-of-function oncogenic mutations in genes encoding the metabolic enzymes IDH1 and, less frequently, IDH2, and thus are genetically distinct from primary glioblastomas1,2. Mutations in IDH1 are commonly found in heterozygosis and often result in the single-amino-acid substitution of arginine (R) with histidine (H) in the catalytic site of IDH1 at codon 132 (called ‘IDH1(R132H)’ here)1,2,3. Mutations in IDH1 and IDH2 result in neomorphic enzymatic activities that lead to production of the oncometabolite 2-HG4,5,6. 2-HG inhibits the enzymatic functions of many α-ketoglutarate-dependent enzymes, including histone and DNA demethlyases, and thus causes the aberrant epigenetic reprogramming seen in the CpG island methylator phenotypes7,8,9,10,11. The presence of mutations in IDH1 and/or IDH2 have led to intensive preclinical and clinical research aimed at developing clinical-grade inhibitors of mutant IDH1, some of which have achieved approval from the US Food and Drug Administration for certain indications, including acute myeloid leukemia or cholangiocarcinoma expressing mutant IDH1; however, despite promising preclinical evidence in support of the efficacy of these compounds, clinical studies of these mutant IDH1–targeted compounds for glioma have not yet progressed beyond early-phase clinical trials, mostly due to concerns about their ability to overcome the blood–brain barrier. This leaves few targeted therapeutic opportunities for patients with glioblastoma, and thus there is an urgent unmet need for the development of alternative strategies that could provide a suitable path for the treatment of these aggressive tumors.

Writing in Nature, Platten and colleagues now report the results of a phase 1 trial testing an anti-cancer vaccine designed to target neoantigens commonly found in patients with glioma bearing IDH1 mutations12 (Fig. 1). Their results provide proof-of-concept evidence of the feasibility and efficacy of this immunotherapy modality and open the path for the development of similar therapeutic approaches for the treatment of these lethal tumors.

figure 1
Fig. 1: Summary of the NOA16 trial.

Anti-cancer vaccination typically consists of the administration of tumor-specific antigens that elicit adaptive anti-tumor immune responses. These antigens usually comprise ‘self’ peptides or ‘non-self’ peptides (such as cancer testis antigens or human papillomavirus proteins, respectively), although the best responses to anti-cancer vaccines are usually achieved when these are directed against antigens expressed exclusively by tumor cells. This has led to greater interest in the use of specific antigens that arise from endogenous tumor mutational processes and are generally known as ‘neoantigens’ or ‘neoepitopes’. Even though anti-cancer vaccines have been extensively explored in various settings with relative success, they have achieved uneven activity in neuro-oncology. More than 70% of diffuse gliomas harbor the IDH1 R132H mutation, an early genetic lesion expressed nearly uniformly by tumor cells, which renders this a potentially powerful therapeutic candidate. Additionally, this neoepitope is presented via the class II major histocompatibility complex (MHC)13 and thus represents an attractive potential target for immunotherapy.

Previous preclinical studies demonstrated that versions of an IDH1(R132H)-specific peptide vaccine (IDH1-vac) were capable of inducing sustained anti-tumor–specific therapeutic helper T cell responses in syngeneic MHC-humanized mice13. Platten and colleagues developed an array of peptides encompassing the R132H substitution within IDH1 and identified the peptide p123–142, which spans the codons 123–142 and includes the R132H substitution, as a potent inducer of specific anti-tumor immune responses to cells expressing mutant IDH1 (Fig. 1). On the basis of this promising preclinical evidence, Platten and colleagues designed a multi-center, phase 1 clinical trial (NOA-16; ClinicalTrials.gov identifier NCT02454634) to test the safety, feasibility and efficacy of a vaccine targeting mutant IDH1 in newly diagnosed patients with World Health Organization (WHO) grade III or grade IV glioma12 (Fig. 1).

In this proof-of-concept trial, Platten and colleagues demonstrated that the IDH1-targeting vaccine was safe and immunogenic and was capable of inducing both T cell and B cell immune responses across patients bearing a variety of human leukocyte antigen–encoding alleles. The authors established a mutation-specificity score to incorporate the duration and level of IDH1-vac-induced T cell immune responses and observed that patients with high scores showed predominant production of the cytokines TNF, IFN-γ and IL-17 by helper T cells, indicative of involvement of the TH1 and TH17 subtypes of helper T cells. The authors also followed up with the patients and assessed the 3-year progression-free and death-free rates, which were 0.63 and 0.84, respectively. Interestingly, treated patients displayed higher rates of pseudoprogression (PsPD), a condition in which patients develop mass lesions that resemble tumor growth by neuroimaging, than those of a molecularly matched cohort that had not been treated with the IDH1-targeting vaccine. PsPD indicates intratumoral inflammatory infiltration and reactions, with prior studies suggesting that patients who develop PsPD after conventional therapy may survive longer. PsPD after vaccination did not correlate with patient age, extent of resection, standard-of-care treatment, or tumor grade. Furthermore, PsPD did not correlate with specific copy-number variations, tumor-methylation class (e.g., the CpG island methylator phenotype), or deletion status for CDKN2A (which encodes a cyclin-dependent kinase inhibitor).

Overall, these results provide evidence that supports the proposal of the induction of specific anti-tumor immune responses after vaccination with a cancer-specific neoepitope.

From the clinic: Kailin Yang and Duane A. Mitchell

Of the 44 patients initially enrolled in the clinical trial, 32 met the inclusion criteria and received the vaccine; the authors were able to perform analysis on the safety and feasibility of this therapy on all the treated patients, including mid-term follow-up, and were also able to perform immunological profiling in a subset of 30 of these treated patients (Fig. 1).

In order to generate a comprehensive map of the complex immunological interactions taking place in the tumor microenvironment following administration of the neoantigen vaccine, the authors performed single-cell RNA sequencing of tumor samples from patients; this allowed them to delineate the heterogeneity of the immune-cell landscape, which eventually resulted in the identification of three distinct clusters of CD4+ T cells within PsPD masses, such as CXCL13+CD4+ T cells, which are key to sustaining anti-tumor immunity responses. Moreover, sequencing of T cell antigen receptors in patients with PsPD revealed that tumor-infiltrating helper T cell clusters displayed a predominance of a single T cell antigen receptor clone, which suggested that IDH1-vac induced the clonal expansion of mutant IDH1–specific helper T cells.

There are notable observations that arise at the interface of the clinical findings and immunological findings of this study that have substantial implications in support of further vaccine development for IDH1-mutant gliomas specifically and for glioma immunotherapy more generally. For example, the observation of vaccine-induced immune responses in more than 90% of the vaccinated participants spanning a broad repertoire of MHC-encoding alleles provides compelling proof-of-concept evidence of the induction of T cell immunity to a highly conserved and ubiquitously expressed glioma-specific mutant protein. Notably, the higher than expected frequency of PsPD in vaccinated participants (and, in a single case, tumor tissue after vaccination) demonstrates an increased frequency of mutant IDH1–specific T cells among tumor-infiltrating lymphocytes.

Despite the promising data in support of the proposal of mutant IDH1 neoepitope–reactive T cells that localize to the central nervous system, further confirmatory studies linking vaccine responses to radiographic and immunological changes are needed. Future prospective studies that incorporate specific imaging endpoints as well as systematic analyses of tumor tissues and/or cerebral spinal fluid will help answer important questions about the potential for imaging modalities to detect changes within gliomas after vaccination; the possibility of monitoring the activity of mutant IDH1–stimulated T cells through liquid biopsy of cerebrospinal fluid; and whether CD4+ T cells that recognize mutant IDH1 effectively elicit anti-tumor activity that translates into improved patient outcomes, or if other populations are involved and required in establishment of the long-term anti-tumor immune responses.

Among the limitations of the study, the trial was not sufficiently powered to determine the relationship between vaccination and patient outcomes, as expected from a phase 1 study; moreover, the overall survival and progression-free survival times for patients with IDH1-mutant gliomas would be expected to be prolonged within this subgroup of patients with glioma. The investigators observed a trend of higher mutation-specificity scores in patients with PsPD than in patients with progressive disease or stable disease, as well as indications of better vaccine-induced responses in patients with stable disease than in those with progressive disease. However, randomized and appropriately powered clinical trials will be needed to determine the long-term impact of this approach, and to discern whether specific immunological correlates can serve as biomarkers of clinical responses. The authors had previously demonstrated that patients have spontaneous T cell and antibody responses to mutant IDH1, and thus it would be very interesting to know whether baseline responses to the mutant protein influence vaccine responses, PsPD and clinical outcomes.

Although the results of this study are encouraging and provide a foundation for further development, some additional outstanding questions remain. For example, during follow-up, four of ten patients with methylation-class high-grade glioma experienced progressive disease. Moreover, PsPD was not detected in patients who did not respond to treatment. Although no molecular correlates were found in this study, it would be interesting to see if future studies can identify patients who will not respond or patients at greater risk of developing progressive disease. Future and larger clinical trials may additionally provide more-granular insights into the various subtypes and stages of this disease by looking at grade III gliomas and grade IV gliomas as distinct entities. As gliomas are highly heterogeneous and develop subclonal alterations during the course of the disease, this vaccine may also provide superior therapeutic indices in combination with other immunotherapy approaches, such as immune-checkpoint inhibition14. As 2-HG has been shown to impair T cell function, small-molecule inhibitors of mutant IDH1 may augment the anti-tumor efficacy of vaccines against mutant IDH115.

As mutant IDH1 is a clonal event, and its expression is detected almost universally in all tumor cells, the vaccine against IDH1 provides the basis of a potentially impactful therapeutic strategy. From a wider point of view, this study—along with the recent scientific advancements in vaccine technology—may provide the foundation for the exploration of vaccine-based therapies for other incurable cancers following a similar rationale, and the work by Platten and colleagues provides the proof-of-concept clinical evidence that will undoubtedly stimulate the development of similarly impactful therapeutic strategies.