Consent Preferences

Elicio’s Therapeutic Vaccine For Pancreatic Cancer: A Revolution Hidden In Plain Sight?

Elicio Therapeutics is developing an off-the-shelf therapeutic mKRAS vaccine for pancreatic cancer. Phase 1 showed 84% T-cell response correlating with prolonged survival. Phase 2 data expected in H1 2026 will determine whether this approach can transform adjuvant therapy.

Elicio’s Therapeutic Vaccine For Pancreatic Cancer: A Revolution Hidden In Plain Sight?
Immunofluorescence staining of PDAC tumor microenvironment: tumor cells (Pan-CK, green), CD4+ T cells (yellow), CD8+ T cells (red), macrophages (CD68, orange), regulatory T cells (FoxP3, blue). Image by the author (© 2025 Chaotropy, all rights reserved).

Disclosure: The author holds beneficial long positions in Elicio Therapeutics (NASDAQ: ELTX) and Revolution Medicines, Inc. (NASDAQ: RVMD) and has no position in BioNTech SE (NASDAQ: BNTX). This article is provided for informational and educational purposes only and is not financial advice. Although the author is a Medical Doctor, this content represents a personal opinion regarding the science and market potential of the companies mentioned and is not medical advice, a diagnosis, or a treatment recommendation. The author neither works at nor has worked at any centers taking part in the studies testing Elicio's candidates and has no involvement in the design, conduct, or analysis of Elicio's clinical trials or scientific programs. The author receives no compensation for this article and has no business relationship with the companies mentioned. Please see the full "Legal Information and Disclosures" section below.

Pancreatic ductal adenocarcinoma (PDAC) remains lethal with an overall 5-year survival of 13% despite decades of research, and is projected to become the second leading cause of cancer death by 2030; no effective targeted therapy has reached the market. Unfortunately, this is where we are now, and at first glance, it is not so different from where we were ten years ago.

Scientists identified the KRAS oncogene as the driver mutation in over 90% of PDAC tumors decades ago, yet targeting it proved far more difficult than drugging cell-surface receptors like HER2 in breast cancer, which present an accessible extracellular domain. KRAS earned its reputation as "undruggable" because the protein lacks the binding pockets that small molecules typically exploit. Its smooth surface, high affinity for intracellular GTP, and location at the cytoplasmic side of the cellular membrane made it impervious to conventional drug design for decades.

This defeatist narrative has now shifted substantially. California-based Revolution Medicines (NASDAQ: RVMD) is leading a wave of direct KRAS inhibitors, with daraxonrasib (RMC-6236) now in Phase 3 trials after demonstrating objective response rates of 35% in RAS G12X and 29% in all RAS mutations and a median progression-free survival of 8.5 months in a heavily pretreated second-line population. In first-line treatment, response rates have reached 47%.

Revolution’s small-molecule approach works through continuous pathway suppression. Daraxonrasib binds to the active form of RAS proteins harboring G12X, G13X, and Q61X mutations, blocking their interaction with downstream effectors. The FDA granted the drug Breakthrough Therapy Designation in June 2025 for previously treated metastatic PDAC with KRAS G12 mutations. Revolution Medicines is now pursuing the adjuvant setting with RASolute 304, targeting patients who have undergone standard of care for resectable PDAC. The rationale is straightforward: if daraxonrasib shrinks metastatic tumors, it should suppress microscopic residual disease.

Continuous dosing comes with costs. In clinical trials, rash occurred in 91% of patients, diarrhea in 48%, and vomiting in 31%. For metastatic patients with limited options, these trade-offs may be acceptable. For adjuvant patients who may already be surgically cured, months or even years of daily pills causing skin eruptions and gastrointestinal distress represent a significant quality-of-life burden. Resistance poses another challenge. Cancer cells adapt: KRAS amplification, pathway reactivation through alternative signaling routes, and secondary mutations inevitably emerge under sustained drug pressure.

Nevertheless, given the platform's potential and wide range of indications (KRAS is mutated in approximately 25% of all solid cancers), Revolution Medicines represents an asset with immense strategic value. If recent market rumors prove accurate, an acquisition by a major pharmaceutical player would be a logical move to secure what could become a backbone of future cancer therapy.

But what if a fundamentally different approach could achieve durable disease control without perpetual treatment?

In clinical practice, I occasionally see PDAC patients in our outpatient clinic who underwent surgery more than five years ago and remain tumor-free. What if the immune system of these long-term survivors holds the key to successful treatment of PDAC? This question motivated researchers to investigate why these patients survived and whether their immune responses could be replicated therapeutically. A landmark study published in Nature in 2017, led by Vinod Balachandran at Memorial Sloan Kettering, examined tumors from long-term PDAC survivors.

The findings were striking. Long-term survivors had 12-fold higher numbers of activated CD8+ T cells infiltrating their tumors than short-term survivors. These T cells were polyclonal, suggesting recognition of multiple tumor antigens. Critically, neoantigen quality, not merely quantity, determined outcomes.

Two competing strategies have emerged to translate this insight into therapy: personalized vaccines that target each patient's unique neoantigens, and off-the-shelf vaccines that target shared driver mutations. BioNTech and Genentech developed autogene cevumeran (BNT122), a personalized mRNA vaccine encoding up to 20 patient-specific neoantigens. In a Phase 1 trial at MSK, the vaccine induced T-cell responses in 50% of treated patients. Responders had dramatically better outcomes: at three-year follow-up, 6 of 8 responders remained disease-free while 7 of 8 non-responders relapsed.

Personalization comes at a cost, as the approach faces practical and economic limitations. Manufacturing an individualized vaccine requires sequencing each patient's tumor, identifying candidate neoantigens, synthesizing custom mRNA, and producing the final product. This process is costly. BioNTech achieved this within nine weeks after surgery, but for a cancer where disease can progress rapidly, this timeline is uncomfortably long. Additionally, only half of the patients mounted the desired immune response, and the Phase 2 trial now underway will determine whether these early signals translate into survival benefits in a larger population. But what if an off-the-shelf alternative could achieve immune activation without the manufacturing complexity?

Boston-based Elicio Therapeutics (NASDAQ: ELTX) is pursuing precisely this strategy. With a market capitalization of approximately $140 million as of Friday, January 23, 2026, which is two orders of magnitude smaller than that of Revolution Medicines, the company is developing ELI-002, an off-the-shelf peptide vaccine targeting the most common KRAS mutations in PDAC. Rather than personalization, Elicio capitalizes on the high prevalence of these specific mutations to create a standardized product that could be administered immediately after surgery, eliminating the weeks-long manufacturing delay and reducing costs.

The company's core innovation is its Amphiphile (AMP) delivery platform. Historical peptide vaccines likely failed because small immunogens injected subcutaneously wash into the bloodstream and are rapidly cleared by the liver and kidneys before reaching lymph nodes, where immune priming occurs.

Elicio solved this by conjugating both the KRAS peptide antigens and the CpG adjuvant to lipophilic tails. These lipid tails bind to albumin, a 67-kilodalton protein too large to cross blood capillary walls. The albumin-bound vaccine complex is instead channeled through lymphatic vessels directly to draining lymph nodes, delivering the immunogen to antigen-presenting cells in the optimal anatomical location.

The AMPLIFY-201 Phase 1 trial enrolled 25 patients with KRAS-mutant cancers (20 PDAC, 5 colorectal) who had completed curative-intent surgery and standard adjuvant chemotherapy but remained positive for Minimal Residual Disease (MRD) based on circulating tumor DNA or elevated tumor markers. Patients received a fixed dose of the Amph-Peptide-2P vaccine targeting KRAS G12D and G12R, alongside escalating doses of the Amph-CpG-7909 adjuvant ranging from 0.1 mg to 10.0 mg. The recommended Phase 2 dose was determined to be 10.0 mg, which was well tolerated and associated with consistent biomarker reductions.

Results published in Nature Medicine in January 2024 reported that 84% of patients developed KRAS-specific T-cell responses, rising to 100% at the highest adjuvant doses (5 mg and 10 mg). According to updated final data published in August 2025, dual priming of both CD4+ and CD8+ T cells occurred in 71% of patients. Among biomarker-evaluable patients, 84% achieved biomarker reductions and 24% achieved complete clearance. High responders demonstrated a 76% median reduction in biomarker levels compared to only 10% in low responders, with 24% achieving complete clearance of ctDNA or CA 19-9/CEA.

The correlation between immune response and clinical outcomes was pronounced. In the final analysis with a median follow-up of 19.7 months, patients with a high T-cell response (defined as above a 9.17-fold increase over baseline) showed a significant divergence in outcomes. The median relapse-free survival for high responders was not reached, compared to just 3.02 months for those with low responses (hazard ratio 0.12; p=0.0002). Similarly, median overall survival was not reached for high responders versus 15.98 months for low responders (hazard ratio 0.23, p=0.0099).

A critic might argue that correlation does not imply causation, pointing to the "healthy user effect." Patients in better health with robust immune systems might respond more strongly to any vaccine, whether for influenza or tetanus, and would likely have better outcomes regardless of the mKRAS vaccine's efficacy.

Fortunately, the authors addressed this potential confounder by testing the T cells of study participants against a panel of infectious antigens like influenza and CMV. The level of these non-specific responses was not significantly associated with biomarker reduction or RFS. In fact, patients with infectious T-cell responses above the median had a numerically worse RFS than those below this threshold (16.33 months vs. not reached; P = 0.102). This suggests that the observed benefit was driven by the specific vaccine mechanism rather than general patient health.

German pharmacologist Gustav Kuschinsky once wrote: "If a substance is said to have no side effects, one must strongly suspect it has no primary effect either." ELI-002 is no exception, as the AMPLIFY-201 Phase 1 trial reported mild side effects: fatigue (19%), headache (19%), and injection site reactions (9.5%). Notably, no dose-limiting toxicities, no treatment-related serious adverse events, and no cytokine release syndrome were observed. In the adjuvant setting, where patients may already be cured, tolerability might determine whether patients complete the regimen and whether the immune system receives sufficient priming to establish durable surveillance.

Phase 2 in the adjuvant setting in PDAC enrolled 144 patients at 24 U.S. sites between January and December 2024 and remains ongoing. While Phase 1 utilized a 2-peptide vaccine, the ongoing Phase 2 study uses an evolved 7-peptide formulation (ELI-002 7P) targeting KRAS G12D, G12R, G12V, G12S, G12A, G12C, and G13D. This broader spectrum covers the most common mutant KRAS subtypes in PDAC, significantly expanding the addressable patient population beyond the G12D/R restriction of the first trial.

The Phase 2 trial compares ELI-002 7P to observation rather than an active comparator, which some investors may view as a lower bar to clear but which is standard practice in the adjuvant setting where no approved therapy exists.

Notably, this means Phase 2 is testing a modified formulation, though the delivery platform and adjuvant mechanism remain identical. At the same time, the Phase 2 study is MRD agnostic, meaning it also enrolls patients without ctDNA or elevated tumor markers after standard of care therapy. This design choice may improve outcomes in both arms but also expands the addressable market if the trial succeeds.

While data from the Phase 2 AMPLIFY-7P trial was expected in 2025, the readout remains delayed. In the company's Q3 2025 financial update (November 2025), Elicio reported the trial was experiencing fewer disease progressions and deaths than projected. An Independent Data Monitoring Committee conducted an "event-driven interim analysis," disclosed in August 2025, and recommended continuation without modification. Elicio further reported that it views this recommendation "as an indication that ELI-002 7P has shown preliminary signals of efficacy," which is a rather positive, albeit cautious, wording. This likely implies an interim hazard ratio favoring the vaccine arm but without statistical significance at the time of analysis.

I would be cautious about interpreting the slower-than-expected event rate as a positive signal for vaccine efficacy. The paradigm shift to neoadjuvant therapy at high-volume centers like MD Anderson and Memorial Sloan Kettering creates a biological selection filter: patients who progress during neoadjuvant chemotherapy never reach surgery and thus never enter the adjuvant trial. Those who do enroll are "biological winners" whose tumors have demonstrated chemotherapy sensitivity. Combined with MRD-agnostic enrollment that includes patients without detectable residual disease, this selection lowers the event rate in both arms, extending the timeline without favoring either group.

Nevertheless, the company expressed cautious optimism in its November 13, 2025, Q3 report, citing the "2:1 randomization" to suggest that the "lower-than-projected number of disease progressions and deaths may reflect a favorable impact on disease-free survival (“DFS”)."

Perhaps the most compelling data was disclosed in December 2025, when Elicio reported that 87% of evaluable Phase 2 patients (13 of 15) treated with ELI-002 7P demonstrated antigen spreading. This indicates that vaccine-induced T cells kill tumor cells expressing the targeted KRAS mutations, releasing additional tumor antigens that are then processed and presented to the immune system, thereby broadening the immune response beyond the original vaccine targets. Antigen spreading suggests the vaccine initiates a cascade of adaptive immunity that leverages the tumor's own mutational repertoire.

Academic interest in the platform is growing. In September 2025, Memorial Sloan Kettering announced an investigator-initiated Phase 1 neoadjuvant trial of ELI-002 7P combined with mFOLFIRINOX and checkpoint inhibition, funded by the Lustgarten Foundation. This expansion into the neoadjuvant setting, where vaccines could prime the immune system before surgical resection, could significantly broaden the addressable market.

Still, Elicio's future depends on the success of the ongoing Phase 2 study, which is far from certain. PDAC is historically a graveyard for initially promising therapies, particularly therapeutic vaccines. Nevertheless, the quality of the science presented by Elicio relative to the current valuation seems asymmetric. Phase 1 data were published in Nature Medicine, a prestigious peer-reviewed journal with rigorous standards. The trial was conducted at leading cancer centers, including MD Anderson and Memorial Sloan Kettering, by investigators with deep expertise in pancreatic cancer immunology. The mechanism of action is biologically plausible and supported by foundational research on long-term survivors. The immunological signals are internally consistent and externally validated by the BioNTech results: the correlation between T-cell response and survival, the biomarker reductions, and the antigen spreading all point cautiously in the same direction.

Crucially, I do not view Revolution Medicines' daraxonrasib as a potential competitor in the adjuvant setting. Continuous pathway suppression and time-limited immune priming address different biological challenges: one maintains constant selective pressure on tumor cells; the other establishes durable immunological surveillance. These approaches may ultimately prove complementary, potentially even synergistic, while serving patients with different tolerance profiles.

As of January 2026, Elicio (NASDAQ: ELTX) remains a speculative micro-cap. In the company's Q3 2025 financial update (November 13, 2025), Elicio reported that "current cash and cash equivalents will support operations through Q2 2026." Although the company currently indicates a study readout for AMPLIFY-7P in H1 2026, there is no guarantee of this. A further delay could lead to further significant dilution or even insolvency.

The company utilizes an at-the-market facility to sell shares directly into the market, creating consistent dilution. The capital structure is further burdened by financing warrants that act as a ceiling on the stock price; as the price rises, institutional holders exercise their options and sell, while the company uses its ATM facility to raise capital, increasing the share count and dampening rallies.

The Phase 2 readout expected in the first half of 2026 represents a binary event. Failure would likely mean insolvency, while success could redefine the standard of care in the adjuvant treatment of PDAC and other solid tumors with KRAS mutations. Given the immense need for novel PDAC therapies, I believe that raising fresh capital, finding a partner for Phase 3, or even a buyout are possible if Phase 2 yields sufficient results.

I currently hold a long position in Elicio based on its asymmetric potential, but I am aware of the risk of total loss of capital. If Elicio succeeds, it would validate not just ELI-002 but the entire premise that lymph node-targeted delivery makes therapeutic off-the-shelf cancer vaccines possible. This would open a new frontier of cancer therapy and start a therapeutic revolution that has been hiding in plain sight.

Follow me on X for frequent updates (@chaotropy).

General Disclaimer & No Financial Advice: The content of this article is for informational, educational, and entertainment purposes only. It represents the personal opinions of the author as of the date of publication and may change without notice. The author is not a registered investment advisor or financial analyst. This content is not intended to be, and shall not be construed as, financial, legal, tax, or investment advice. It does not constitute a personal recommendation or an assessment of suitability for any specific investor. Readers should conduct their own independent due diligence and consult with a certified financial professional before making any investment decisions.

Medical Disclaimer: Although the author possesses a medical background, the information presented here regarding medical technologies, clinical trials, or pharmaceutical mechanisms is strictly for the purpose of educational discussion and general commentary regarding the underlying science. It does not constitute medical advice, diagnosis, or treatment recommendations, nor does it establish a physician-patient relationship. Readers should never disregard professional medical advice or delay in seeking it because of something read on this website. Always consult a qualified healthcare provider regarding any medical condition.

Accuracy and Third-Party Data: Market trends, clinical trial data, and performance metrics referenced in this article are sourced from independent third parties. While the author believes these sources to be reliable, the completeness, timeliness, or correctness of this data cannot be guaranteed. The author assumes no liability for errors, omissions, or the results obtained from the use of this information.

Disclosure of Interest: The author holds beneficial long positions in Elicio Therapeutics (NASDAQ: ELTX) and Revolution Medicines, Inc. (NASDAQ: RVMD). The author does not hold any position in BioNTech SE (NASDAQ: BNTX). The author reserves the right to buy or sell these securities at any time without further notice. The author receives no direct compensation for the production of this content and maintains no business relationship with the companies mentioned.

Independence from Elicio Therapeutics: The author does not work at nor has worked at any centers participating in the studies testing Elicio's candidates. The author has no involvement in the design, conduct, funding, data collection, analysis, or interpretation of Elicio Therapeutics' clinical trials or scientific programs. All opinions expressed regarding Elicio are based solely on publicly available information.

Forward-Looking Statements & Risk: This article contains forward-looking statements regarding regulatory outcomes (such as FDA decisions), clinical results, and market potential. These statements are predictions based on current expectations and are subject to significant risks and uncertainties. Investing in biotechnology and pharmaceutical securities involves a high degree of risk, including the potential for total loss of principal. Past performance is not indicative of future results.

Copyright: All content is the property of the author. This article may not be copied, reproduced, or published, in whole or in part, without the author's prior written consent.

Do Not Sell or Share My Personal information