Entrepreneurship

iGEM Criteria

Below is a concise overview of how Oncoligo meets iGEM’s entrepreneurship criteria. We invite you to explore our full entrepreneurship content for a complete view of our strategy, progress, and long-term vision.

Mission Statement

Problem Focus

Cancer remains one of the greatest challenges facing modern medicine, responsible for millions of deaths each year worldwide - a number projected to keep rising. Despite being a collection of diverse diseases, all cancers share a core mechanism: genetic mutations that cause cells to behave abnormally and aggressively. The close similarity between cancerous and healthy cells makes effective treatment particularly difficult. Conventional therapies such as chemotherapy and radiation often damage healthy tissue, resulting in severe side effects and limited long-term success. Moreover, these approaches may fail to eliminate all cancer cells, leading to recurrence. In recent years, oncology has begun to embrace precision medicine - targeted therapies that aim to treat each patient according to their specific genetic and molecular profile. However, current options still lack the specificity and flexibility needed to overcome many of cancer’s complexities.

Despite decades of research, today’s cancer treatments remain limited in several critical ways. Most therapies address only a narrow subset of mutations, leaving many cancer-driving changes untargeted. Others focus on a single pathway, giving tumors the opportunity to escape through alternative mechanisms. Effective delivery into tumor cells continues to be one of oncology’s greatest hurdles, and even when treatment succeeds initially, no drug is able to eliminate all malignant cells, allowing relapse to occur. Above all, therapies must be both precise and adaptable to the extraordinary diversity of cancer - a standard that current approaches still fail to meet.

This gap is especially urgent in lung cancer, one of the deadliest malignancies worldwide, where existing therapies often prove insufficient and long-term outcomes remain poor. Together, these challenges – the inability to address diverse mutations, the reliance on single pathways, the lack of reliable delivery methods, and the failure to achieve complete tumor elimination- show why incremental improvements are no longer enough. What is needed is not another variation on existing therapies but a fundamentally new paradigm: one that unites precision, adaptability, effective delivery, and long-term protection against relapse. To overcome these barriers, a fundamentally new approach to cancer therapy is required -and this is exactly the mission of ONCOLIGO.

Our Mission

ONCOLIGO’s mission is to redefine the way cancer is treated by uniting safety, precision, and adaptability in one therapeutic platform. Unlike existing approaches that are restricted to a narrow set of targets, we are committed to creating treatments that can address a broad spectrum of cancer-driving mutations. By exploiting the concept of synthetic vulnerabilities, our platform is designed to strike multiple weaknesses at once, reducing the chance of tumor escape.

One of oncology’s greatest bottlenecks has always been ensuring that therapies effectively reach tumor cells. Our approach therefore places targeted delivery at its core, with lung cancer chosen as the proof-of-concept indication. To guarantee accuracy and flexibility, the platform is built to be computationally driven, modular, and rapidly adaptable to different cancers. And because no single therapy can eliminate all malignant cells, ONCOLIGO also incorporates mechanisms to engage the body’s natural defenses and achieve more durable responses.

Our mission is therefore not only to create new drugs, but to establish a versatile and expandable platform that closes long-standing gaps in oncology – from mutation coverage and tumor targeting to relapse prevention. Our ultimate vision is to provide patients and clinicians with next-generation precision oncology solutions that minimize harm, maximize long-term efficacy, and reshape the future of cancer care.

To capture the essence of our mission at a glance, we summarize it in three simple questions - Why, What, and How:

Why, What, and How
Figure 1: ONCOLIGO’s mission summarized through Why, What, and How - the need, the solution, and the mechanism.

Unmet Need in Lung Cancer Treatment

Lung cancer remains one of the most challenging and deadly forms of cancer, with many patients still lacking effective, long-term treatment options. In this section, we outline the urgent unmet need by highlighting three key aspects: the global burden and rising incidence of the disease, the critical limitations of current treatment modalities, and the firsthand experiences of both clinicians and patients confronting these gaps in care.

Disease Burden

Lung cancer remains one of the leading causes of cancer-related mortality worldwide. According to the American Cancer Society 2025 report, lung cancer is responsible for approximately 1 in 5 cancer deaths, with an estimated 226,650 new cases and 124,730 deaths (Lung & bronchus cancer) expected in the United States alone this year [1]. Globally, according to GLOBOCAN 2022, lung cancer was the most commonly diagnosed cancer, accounting for nearly 2.5 million new cases (12.4% of all cancers), and the leading cause of cancer death, responsible for 18.7% of global cancer-related mortality. The burden is particularly high among men, though incidence among women is rising, with substantial geographic variation observed across continents [2].

The World Health Organization (WHO) attributes the rising burden of lung cancer, particularly in regions such as Asia, to increasing tobacco consumption [3].

Age-standardized incidence of lung cancer worldwide in 2022
Figure 2: Age-standardized incidence of lung cancer worldwide in 2022 [2]

Despite advances in targeted therapies and immunotherapies, the five-year survival rate for lung cancer remains below 30%, largely due to late-stage diagnosis and limited treatment options for patients with rare or less-studied mutations [1].

Five-year relative survival rates for selected cancers by race and stage at diagnosis, United States, 2014 to 2020
Figure 3: Five-year relative survival rates for selected cancers by race and stage at diagnosis, United States, 2014 to 2020 [1]

According to GLOBOCAN 2022, the number of new cancer cases worldwide is expected to reach over 35 million by 2050, marking a 77% increase compared to approximately 20 million cases reported in 2022. This projected rise is largely attributed to population growth and ageing, occurring alongside a global demographic shift toward lower birth and death rates. In parallel, the International Agency for Research on Cancer (IARC) anticipates a significant rise in lung cancer, with incidence projected to grow by 58.8% and mortality by 64% between 2020 and 2040 [2].

The development of lung cancer is strongly associated with environmental and behavioral risk factors. Smoking remains the predominant cause, contributing to approximately 85–90% of all cases. However, other exposures, such as second-hand smoke, family history, and contact with carcinogenic substances are also significant contributors. Additionally, underlying medical conditions like pulmonary fibrosis, HIV infection, and lifestyle factors such as alcohol use further increase the likelihood of developing the disease [3].

Despite advances in surgery, radiation, chemotherapy, and immunotherapy, non-small cell lung cancer (NSCLC) remains a major clinical challenge, particularly due to the high rates of recurrence and metastasis after treatment. NSCLC accounts for approximately 85% of all lung cancer cases, with three main types - adenocarcinoma (40%), squamous cell carcinoma (25–30%), and large cell carcinoma (5–10%). However, many patients who undergo curative-intent surgery still experience distant metastases or local recurrence, highlighting the limitations of current therapeutic strategies and the urgent need for novel approaches [3].

NSCLC is often diagnosed at an advanced stage, limiting treatment options. The most common symptom is cough (50–75% of cases), but its nonspecific nature contributes to delayed detection. Despite the use of PET-CT and liquid biopsy, accurate diagnosis still requires tissue sampling [3].

Metastatic non-small cell lung cancer (mNSCLC) imposes a substantial economic burden on the U.S. healthcare system, particularly during the period between diagnosis and the initiation of first-line treatment. A 2023 ISPOR study revealed that for patients with commercial insurance, average per-patient-per-month (PPPM) costs during this early phase reached \$64,253, driven primarily by medical services (\$61,824) and pharmacy-related expenses (\$2,429). Even for patients with public insurance (Medicaid), PPPM costs exceeded \$34,000. These costs decline in subsequent phases, but remain significant. Notably, patients with mNSCLC without actionable EGFR or ALK mutations, who make up the majority, face limited treatment options despite repeated healthcare utilization and high failure rates of existing first-line therapies. This gap highlights both a clinical and financial need for a new biologic therapy that accelerates therapeutic efficacy or delays progression. Such an intervention could reduce resource-intensive care periods and hospitalization rates, lowering payer expenditures while improving patient outcomes [4].

Our project focuses on developing an antisense oligonucleotide (ASO) based therapy tailored to a genetically defined subpopulation of lung cancer patients. To ensure both clinical relevance and scalability, we aim to target a mutation (or set of mutations) present in at least 5% of lung cancer patients. This precision approach will allow us to address a biologically significant subset of patients who may not respond well to current treatments.

By identifying such a subgroup and designing molecular therapy, we aim to fill a critical gap in personalized oncology. Our approach is particularly relevant for non-small cell lung cancer (NSCLC), which accounts for about 85% of all lung cancer cases, and where genetic heterogeneity poses a challenge for effective, long-term treatment.

Current Treatment Limitations

Despite major advances in therapy over the past two decades, significant limitations persist across existing treatment modalities.

Traditional chemotherapy kills rapidly dividing cells indiscriminately, leading to severe side effects and often incomplete tumor eradication [5], the last two decades have seen the rise and consolidation of all major cancer treatment modalities. These include targeted therapies (e.g., EGFR, ALK inhibitors) and biological drugs (e.g., monoclonal antibodies like Nivolumab or Bevacizumab), which act more selectively on cancer-driving molecules [6],[7]. At the same time, traditional treatments such as chemotherapy and radiotherapy remain widely used in clinical practice, although the primary focus of research and innovation has shifted toward more precise and personalized approaches.

These newer therapies offer improved safety and efficacy - but only for a subset of patients with specific genetic mutations or protein biomarkers, and even these advanced therapies have limitations:

  1. Restricted to Membrane: Most targeted therapies act on cell-surface proteins. This restricts their utility because intracellular targets, such as mutated proteins, mRNA, or non-coding RNAs, remain largely inaccessible. Antibody development is limited to extracellular targets and is expensive, time-consuming, and often hampered by tumor heterogeneity and antigen loss [8],[9].
  2. Mutation-Driven Resistance: Cancer cells are highly dynamic. During treatment, secondary mutations, alternative splicing, or gene amplifications can emerge, rendering once-effective drugs obsolete. This is especially common with EGFR and ALK inhibitors, where resistance often develops within months. As resistance accumulates and toxicities limit further treatment, some patients exhaust all available drug options, leaving their physicians with no additional therapies to offer - a devastating outcome that underscores the need for more versatile and durable treatment strategies [10],[11].
  3. Patient Eligibility Is Narrow: Precision oncology often depends on identifying specific mutations or biomarkers in each patient. Even within a single gene, such as EGFR or KRAS, dozens of distinct mutations can exist - each potentially requiring a different therapeutic approach. As a result, most drugs are only effective for a small subset of patients with the "right" mutation, leaving many without an applicable targeted treatment [12].
  4. Missed Opportunities: Targeted therapies have largely focused on oncogenic drivers - genes that gain function and can be inhibited. In contrast, tumor suppressor genes such as TP53, RB1, and APC, which are inactivated in many lung cancers, present a major therapeutic gap. Their loss-of-function nature, combined with the diversity of mutations involved, makes it nearly impossible to "repair" them with a single drug. As a result, these critical genes are often ignored in current treatment paradigms.

To better understand the existing landscape, we compiled a comprehensive table of all FDA-approved non-chemotherapy treatments for lung cancer - including targeted therapies, immune checkpoint inhibitors, and angiogenesis inhibitors.

See Table: “Lung Cancer non-chemotherapy Current Approved Therapies”

While modern therapies have improved outcomes for select patient populations, the limitations outlined above reveal substantial gaps in lung cancer treatment - particularly for patients lacking druggable oncogenic drivers or harboring tumor suppressor gene mutations. These challenges highlight the urgent need for novel, versatile approaches that can overcome resistance, broaden patient eligibility, and effectively target the currently “undruggable” vulnerabilities in cancer.

Patient/Clinician Insight

While lung cancer remains the leading cause of cancer-related death worldwide, the personal experiences of patients and clinicians reveal a deeper, more human dimension of this crisis. One defined not only by biology, but by pain, limitations, and unmet therapeutic needs.

During our meeting with Prof. Amir Onn, Chair of the Institute of Pulmonary Oncology at Sheba Medical Center, he shared the story of a young mother he has been treating since 2016. After nine years of battling lung cancer, none of the available treatments are effective anymore. “I have nothing left to give her”, he said. “No drug works anymore”.

This sense of helplessness, echoed by clinicians facing treatment resistance, is deeply connected to the lived experiences of patients. As therapies lose efficacy over time, patients are left struggling not only with physical decline but with the emotional weight of limited options and uncertain futures.

A 25-year-old nurse shared her story after being diagnosed with stage IV non-small cell lung cancer despite having no risk factors. She recalled the emotional toll of chemotherapy [13]:
“I started chemotherapy and fell into a deep depression. One day, in between chemo sessions, I remember sitting in my bed and thinking: "It would be so much easier if I just ended it now and saved everybody the trouble.”

Following initial chemotherapy, she began drug therapy that brought temporary improvement, but she remained aware of its limitations:
“Drug therapy isn't a cure; it’s a treatment. I had to come to terms with that. Eventually, the drugs will become less effective as my body adapts and the cancer finds ways around it.”

Still, she expressed cautious hope for the future:
“The hope is to one day treat this type of lung cancer, or all cancers, like we treat diabetes—as a chronic condition that can be managed.”

These personal accounts are echoed by the study 'Symptoms and experiences of frailty in lung cancer patients with chemotherapy' [14] which explored the experiences of 302 lung cancer patients undergoing chemotherapy. Many reported severe fatigue, appetite loss, anxiety, and social withdrawal.

Further supporting this picture, the study 'Living with advanced or metastatic lung cancer - A qualitative study on the experiences of patients' [15], interviewed 19 patients with advanced or metastatic NSCLC. Participants described how symptoms like shortness of breath, persistent cough, and fatigue interfered with walking, sleeping, and daily life. They also reported confusion about whether their symptoms came from the disease or the treatment - adding emotional distress to physical discomfort.

In addition to published reports, our interviews with patients shed further light on their challenges and needs. Ellen Nemetz, a 64-year-old artist from Arizona diagnosed with stage IV lung cancer carrying the rare KRAS-Q61H mutation, shared her experience with cisplatin, Pemetrexed, and Keytruda. She described cisplatin as “the most difficult chemotherapy, "with severe fatigue and constipation, but emphasized how physical activity helped her cope. Ellen highlighted the urgent need for therapies tailored to rare mutations, stressing that “patients need treatments that consider quality of life as much as efficacy."

We also spoke with a patient who preferred to remain anonymous (“S”), living with EGFR-mutant lung cancer. After an initially dramatic response to targeted therapy (dacomitinib), her disease progressed and she transitioned to chemotherapy, which she described as "a completely different world," marked by debilitating fatigue and cognitive fog. Beyond the medical toll, she pointed to the bureaucratic and financial struggles of navigating insurance and drug access, emphasizing that "beyond efficacy, it is critical to reduce toxicity, simplify access, and design solutions that are realistic for patients managing complex, long-term disease."

Together, these insights highlight the urgent need for treatments that reduce systemic toxicity, preserve patients’ quality of life, and adapt to the evolving biology of lung cancer. ASO-based therapeutics represent a promising direction toward fulfilling this unmet need.

Solution and Tech

To address the key limitations highlighted in our mission statement, ONCOLIGO introduces a tri-functional therapeutic platform that unites precision, safety, and adaptability in one system. This platform is designed to overcome the difficulty of treating diverse mutations, the challenge of treatment resistance, the barriers to effective delivery, and the need for durable and accurate responses.

Having established lung cancer as our initial focus due to its urgent unmet need, we now present how ONCOLIGO’s platform is built to directly respond to these challenges and demonstrate its potential as a broadly adaptable solution.

Core Solution

At its core, ONCOLIGO is built on a programmable antisense oligonucleotide (ASO) designed to silence cancer-driving RNA. The ASO is optimized by a computational model that integrates a broad spectrum of biological and biophysical features -including RNA structure, hybridization dynamics, accessibility, codon usage, nuclease sensitivity, and off-target potential to generate sequences that are both highly specific and effective.

A central strength of the platform lies in its use of synthetic lethality: in tumors where a tumor suppressor gene is lost or mutated, ONCOLIGO targets its druggable synthetic lethal partners. This approach ensures selective killing of cancer cells while leaving healthy tissue intact, opening the door to treating cancers that were previously considered "undruggable", such as, silence or loss of function mutations.

Safety is further strengthened by the BROTHERS system, a molecular safety switch that activates the ASO only when the correct RNA target is detected [16]. To address one of oncology’s most persistent barriers - effective delivery - ONCOLIGO employs antibody-based targeting of tumor-specific surface markers. Once inside the tumor cell, peptide epitopes derived from cancer or pathogens enhance antigen presentation and stimulate cytotoxic T-cell responses. As therapies may not reach or eradicate every tumor cell, engaging the immune system is crucial to help eliminate residual disease and provide durable protection against relapse.

Figure 4: Schematic illustration of the ONCOLIGO therapeutic construct - showing how the ASO is coupled with an antibody for targeted delivery, secured by the BROTHERS safety switch, and enhanced with epitopes to stimulate immune activation.

As an initial proof of concept, ONCOLIGO is being developed for a genetically defined subset of lung cancer patients: non-small cell lung cancer (NSCLC) tumors with MTAP deletions, representing approximately 16% of patients based on cBioPortal datasets [17]. The first application combines a computationally designed ASO directed against MTAP synthetic-lethality partners such as PRMT5, RIOK1, and MAT2A [18]. In addition, based on our own analyses (detailed in the Target Selection section), we are also evaluating loss-of-function mutations in TP53 and their synthetic-lethality partners, including TP53BP1 and RB1, as potential targets [18].

Key Aspects of the Platform

While our initial focus in lung cancer illustrates how ONCOLIGO can be applied to a defined patient population, the true strength of the platform lies in its complementary layers. Each layer was designed to address a specific weakness of current cancer therapies, and together they create a multi-functional system where every component is essential. The table below outlines each aspect, explaining the clinical need it answers, the solution we propose, and the value it creates.

Aspect Illustration Need Solution Value
ASO Current drugs cannot address many cancer-driving mutations, especially those that do not produce proteins. ASOs silence RNA directly, enabling broad mutation coverage Unlocks new therapeutic markets by targeting patient populations previously left without options.
Antibody Most RNA therapies fail because they cannot reach tumor cells effectively. ONCOLIGO uses antibody-guided delivery specific to lung tissue to ensure uptake where it matters. Overcomes one of the biggest translational barriers in oncology, enabling real-world clinical adoption.
BROTHERS Off-target effects remain a critical barrier to RNA therapeutics. The BROTHERS safety switch activates the ASO only in the presence of the correct RNA target. Differentiates ONCOLIGO as a safer therapy, reducing regulatory risk and increasing patient trust.
Epitopes Even effective therapies often leave behind residual tumor cells, leading to relapse. ONCOLIGO incorporates tumor- and pathogen-derived epitopes to stimulate the immune system for durable clearance. Extends patient survival and strengthens the therapeutic profile, making it attractive in competitive oncology markets.
Computational Model Drug design in oncology is often slow and case-specific, limiting scalability. ONCOLIGO's proprietary computational pipeline ensures accurate, modular, and rapid ASO design. Provides flexibility to design therapies for diverse cancer-driving mutations and scalability across multiple cancer types, positioning ONCOLIGO to deliver both a concrete initial product and a broader adaptable platform for oncology.
Synthetic Lethality Many tumor suppressor mutations are "undruggable," and directly restoring their function is impractical. we need a way to exploit cancer-specific dependencies without harming normal cell Leverage synthetic lethality: in tumors harboring a loss-of-function in Gene A (TSG), therapeutically inhibit its druggable partner Gene B so that the combined dysfunction (A mutated, B inhibited) triggers selective cancer cell death, while normal cells with intact A tolerate B inhibition. Enables treatment of "undruggable" drivers by acting on druggable partners; provides coverage across heterogeneous mutations that converge on the same SL partner; improves therapeutic index and supports durable responses by targeting tumor-specific dependencies.
Figure 5: summary of ONCOLIGO’s key aspects. Each row highlights a clinical need, the solution provided by the platform, and the value it creates , accompanied by an illustration of the therapeutic component.

While other approaches may address some of these elements, ONCOLIGO uniquely integrates them into one coherent system. Together, these innovations transform ONCOLIGO from a single therapeutic candidate into a true platform that can expand across multiple cancer types. By combining precision, safety, delivery, immune activation, and computational design, ONCOLIGO becomes a safe, adaptable, and impactful therapeutic solution. To learn more about the solution and technology mechanisms see the Description section.

This integration defines ONCOLIGO’s uniqueness and prepares the ground for understanding how our approach compares with existing solutions in the oncology landscape -a topic explored in the next section.

Competitive Landscape & Differentiation

RNA therapeutics are rapidly transforming oncology, with numerous companies developing oligonucleotide approaches [19],[20]. Our iGEM project pushes the boundaries by integrating three synergistic strategies:

  • Antisense Oligonucleotide (ASO) generated by our unique computational model. designed either to silence synthetic lethality targets or to modulate splicing for neoantigen creation.
  • Antibody Conjugation for precise tissue targeting, optimized with multiple computational tools and models.
  • Epitope Conjugation to recruit and activate T cell cytotoxicity, derived from smart computational pipeline.

This multi-component, modular combination addresses intracellular mutations and boosts immune-mediated tumor killing - an innovation no competitor currently offers for lung cancer. Below, we analyze the competitive landscape and explain why our approach offers significant advantages.

What Makes Oncoligo Unique?

Feature Our Platform Other Players
Modality ASO + Antibody + Epitope RNA or Antibody - not both
Modularity and flexibility
Each component can be swapped or redesigned: the ASO sequence can target a new gene, the antibody can be optimized differently and replaced to fit different cancer tissues, and the epitope can be changed and adapted to engage alternative T cell responses. Non-modular – therapies are fixed designs with little to no flexibility
Target
Intracellular mRNA Primarily target surface proteins or mRNA for degradation only; none focus on splicing modulation or synthetic lethality partners of tumor suppressor genes.
Mechanism
Synthetic lethality, mRNA knockdown, Splicing modulation and immune system activation. Protein inhibition or knockdown only
Delivery
Antibody-guided intracellular delivery Some rely on lipid/viral carriers (less specific), while others have not yet developed clear delivery strategies
Computational Model
Our advanced modeling pipeline predicts RNA accessibility, splicing impact, ASO efficiency, off-target probability, optimal chemical modifications, antibody optimization, epitope conjugation, and many more features. This ensures rational design rather than trial-and-error (See our Model page for details). Rely on conventional design methods with limited prediction of RNA accessibility or splicing effects. Often depend on empirical screening and trial-and-error, leading to slower optimization and less precise ASO candidates.

We divide our competitors into direct, indirect and Replacement competitors.

We created a table summarizing current leading direct and indirect competitors companies - (See: “Direct & indirect Competitors”), and replacement competitors - (See: “Replacement competitors”).

Direct Competitors - RNA-Based Cancer Therapies

These companies develop therapies that silence gene expression with RNA tools, but do not use antibody or epitope conjugation. Example companies:

Company Cancer Type Modality Gene Target
Flamingo Therapeutics Head & Neck SCC, AML/MDS ASO STAT3
Flamingo Therapeutics Triple Negative Breast Cancer ASO MALAT1
HAYA Therapeutics Solid Tumor Microenvironment ASO Undisclosed
Secarna Pharmaceuticals Solid Tumors ASO SECN-15
Sirnaomics Multiple Solid Tumors siRNA TGF-β1, COX-2
Adarx (with Abbvie) Undisclosed siRNA Undisclosed
Phio Pharmaceuticals Melanoma, Merkel Cell Carcinoma siRNA PD-1
TransCode Therapeutics Various siRNA MYC, PD-L1

In particular, siRNA/shRNA-based therapies, though powerful, have key limitations:

  • Depend on RISC complex saturation
  • Act only in the cytoplasm
  • Are less stable
  • Cannot modulate splicing
  • Have higher mismatch tolerance, increasing off-target effects [21]
  • Are harder to deliver due to their double-stranded nature [21]

In contrast, ASOs:

  • Work via RNase H or steric blocking and splicing modular
  • Can act in both nucleus and cytoplasm
  • Are simpler to synthesize and modify chemically

By leveraging synthetic lethality, our approach can selectively kill cancer cells harboring specific mutations while sparing healthy cells - something no competitor currently implements in lung cancer with ASOs. By targeting the synthetic lethality partner rather than each individual mutation, we gain the major advantage of addressing a wide spectrum of tumor mutations through a single therapeutic strategy (see figure below).

Synthetic Lethality Strategy
Figure 6: Illustration of synthetic lethality strategy. Multiple mutations across the tumor suppressor gene (top) are represented by red blocks, each affecting different patients. Instead of targeting each mutation separately, synthetic lethality targets the partner gene (bottom), enabling a single therapeutic approach to address all mutations simultaneously.

Indirect Competitors - Antibody-Conjugated Therapies

These companies develop antibody-drug conjugates (ADC) or immune engager antibodies. They deliver toxins, proteins, or immune activators, but not RNA. Example companies:

Company Cancer Type Modality Gene Target
LigaChem Biosciences HSolid Tumors ADC Smart linker + toxin
CytomX Therapeutics Solid Tumors Probody ADC Conditional activation
TrojanBio Solid Tumors Antibody–Epitope Conjugate Immune activation

Replacement Competitors - Standard-of-Care Lung Cancer Therapies

Patients today rely on standard-of-care therapies (non-RNA):
Small molecule inhibitors: EGFR, ALK, KRAS G12C (e.g., Sotorasib)
Immunotherapies: Anti-PD-1/PD-L1 (Nivolumab, Pembrolizumab)
Cell therapies: CAR-T

Key gap: Often limited to narrow patient subgroups, develop resistance, and do not target intracellular RNAs.

Company/Product Cancer Type Modality Target/Mechanism
Amgen (Sotorasib) NSCLC (KRAS G12C) Small molecule inhibitor KRAS G12C
AstraZeneca (Tagrisso) NSCLC (EGFR) Small molecule inhibitor EGFR
Merck (Keytruda) NSCLC Immunotherapy PD-1
Bristol Myers Squibb (Opdivo) NSCLC Immunotherapy PD-1
LYL797 (Lyell Immunopharma) TNBC, NSCLC CAR-T T cell reprogramming

While these are clinically approved or in development, they primarily:

  • Target membrane proteins, leaving intracellular RNAs untargeted
  • Are effective only in narrow patient subsets
  • Lose efficacy over time due to mutations or pathway redundancy

Future Competitors and Our Strategy to Stay Ahead

We expect future competition from:

  • Pharma companies expanding ASO-based therapies for lung cancer, specifically targeting synthetic lethality pathways similar to ours.
  • ASO + Antibody combinations, as big pharma and biotech look to integrate RNA payloads into antibody platforms to improve targeting and delivery.
  • Epitope-ASO conjugates or new technologies that use ASOs to generate neoantigens, activating the immune system to kill tumor cells with patient-specific T cell responses.

Our strategy to maintain a competitive edge relies on three main pillars:

1. Continuous Innovation Through Modularity

Our modular platform allows rapid swapping or redesign of each component - ASO sequences, antibody carriers, and epitope payloads - without rebuilding the entire system. This agility ensures:

  • Fast adaptation to new cancer targets: If competitors develop similar ASOs, we can immediately retarget novel synthetic lethality partners or splice sites.
  • Flexible delivery solutions: Antibody or epitope components can be replaced with improved carriers as technology advances.

2. Early Clinical and Partnership Strategy

We plan to:

  • Collaborate with pharma companies for delivery optimization, GMP manufacturing, and early-phase clinical trials.
  • Engage regulatory agencies early to establish clear translational pathways, securing first-mover advantage in synthetic lethality–based RNA therapeutics for lung cancer.

3. Intellectual Property (IP) Protection

As detailed on our Regulatory & IP Strategy, we patent ASO sequences, epitope constructs, antibody designs, modular platform topology, and computational algorithms. This multi-level patenting ensures that even if competitors develop similar concepts, they cannot replicate our specific sequences, designs, or methods without infringing.

4. Broad Clinical Target Space

Unlike therapies limited to single mutations or pathways, our synthetic lethality approach covers multiple mutations simultaneously. Even if competitors emerge, we can expand rapidly to new cancer subtypes and resistance mechanisms.

Competitive Analysis Tablr
Figure 7: Comparative analysis highlighting Oncoligo’s unique position in the RNA therapeutics landscape.

Since our platform is multi-component, we created several positioning maps(Figures 1-3). Each one highlights a different dimension of our advantages compared to competitors, showing how Oncoligo outperforms.

Competitive Analysis Matrix
Figure 8: Competitive Analysis Matrix

This matrix compares leading companies in RNA therapeutics along two key dimensions:

  • Resistance Risk (Y-axis): Likelihood of therapeutic resistance development.
  • Intracellular Targeting Depth (X-axis): Ability to achieve deep intracellular delivery and targeting.

Oncoligo shows strong performance in Intracellular Targeting Depth with low Resistance Risk, while competitors vary in position across both axes.

Competitive Analysis Matrix
Figure 9: Competitive Analysis Matrix - Modularity vs. Computational Design Capability

This matrix compares companies in RNA therapeutics across two dimensions:

  • Modularity (Y-axis): Flexibility of the platform to incorporate multiple therapeutic components.
  • Computational Design Capability (X-axis): Integration of computational modeling and design in therapeutic development.

Oncoligo leads with strong Computational Design Capability and Modularity, while competitors show varying strengths across both axes.

Competitive Analysis Matrix
Figure 10: Competitive Analysis Matrix – Functionality vs. Delivery Efficacy

This matrix compares companies in RNA therapeutics along two key parameters:

  • Functionality (Y-axis): Breadth and versatility of therapeutic mechanisms.
  • Delivery Efficacy (X-axis): Efficiency of intracellular delivery and therapeutic performance.

Oncoligo ranks highest in Delivery Efficacy and Functionality, with competitors distributed across both dimensions.

Product - SWOT Analysis

Strong strategy starts with clear insight. This SWOT analysis maps Oncoligo’s strengths, weaknesses, opportunities, and threats-showing how we learn, adapt, and build toward a stronger future. Explore the full overview in the PDF below.

Market Analysis

When developing a therapeutic solution, it is important to understand not only the scientific and technical feasibility, but also the potential impact in the healthcare market. To evaluate this, we use the concepts of TAM, SAM, and SOM, which help define the scope of opportunity for our project.

Having defined these concepts, we next applied them to our project in order to estimate the market potential:

How did we calculate the TAM-SAM-SOM?
(Bottom-Up approach)

As we create a new drug that has not existed before, aimed at patients without effective treatments, we are effectively creating a new market. A bottom-up approach - extrapolating from the cost of treating a single patient - is therefore appropriate.

SAM (US, MTAP-deleted NSCLC patients):

In 2025, ~226,650 people are projected to be diagnosed with lung cancer in the US [22]. About 85% have NSCLC, and ~15% are MTAP-deletion-positive [23], yielding ~29,000 patients annually. At a price of \$176,000 per patient per year, this implies a US market size of ~\$5.1B.

SOM (US, MTAP-deleted NSCLC patients eligible for our therapy):

Of these ~29,000 patients, an estimated 20–40% have no targetable molecular alterations [24]. Since MTAP deletions are distributed across mutations [25], we can conservatively assume ~20% of the 29,000 patients represent the true addressable pool. This yields ~5,800 patients, or ~\$1.02B at US pricing.

TAM (Global MTAP-deleted lung cancers):

In 2022, 2.48M people were diagnosed with lung cancer worldwide [26]; outside the US this corresponds to ~2.25M. With ~85% NSCLC, that is ~1.91M patients. Applying a 15% MTAP-deletion rate gives ~286,000 patients. Cancer drug prices outside the US are typically discounted; European prices are ~52% lower [27]. Conservatively assuming a 75% discount yields ~\$12.6B globally. Adding the US SAM, total TAM is ~\$17.7B.

Clarifying Notes

  • Lower bound assumption: We estimate that the percentage of MTAP deletion in SCLC is low and approximate it as zero. Actual numbers may be higher.
  • Conservatism: These figures are conservative estimates, applying multiple narrowing filters to avoid overestimation.

Beachhead Market

Our beachhead market represents the first group of patients who would directly benefit from our therapy and form the foundation for clinical adoption. These are U.S. patients with non-small cell lung cancer (NSCLC) who are MTAP-deletion–positive and do not have effective targeted drugs available.

This subgroup is especially critical because:

  • High unmet need - These patients currently have limited or no targeted treatment options, relying largely on chemotherapy or non-specific immunotherapies.
  • Clear clinical identification - MTAP deletion can be reliably detected using existing genomic panels, enabling straightforward patient stratification.
  • Sizeable and accessible market - With ~29,000 U.S. MTAP-deleted NSCLC cases per year, and ~20% meeting our eligibility assumptions, this translates to ~5,800 patients annually.
US map with lungs

To address this need, our Minimum Viable Product (MVP) is a MTAP-targeted ASO-antibody complex specifically designed for this patient population. This targeted therapy aims to exploit the MTAP deletion vulnerability, offering a novel and more precise treatment alternative.

Focusing on this population allows us to demonstrate clinical efficacy, establish safety, and build early market traction before expanding into broader NSCLC populations and eventually global markets. In other words, this beachhead is not just our first commercial opportunity - it is the strategic entry point that validates our therapy and opens the path to scale.

Business Model & Value Proposition

Every great company starts with a vision. This business model shows how Oncoligo transforms a scientific breakthrough into a sustainable venture-bridging discovery, strategy, and impact. Discover how we plan to turn vision into reality in the PDF below.

Pitch

Every breakthrough begins with a story. In this pitch video, we invite you into Oncoligo’s journey- from identifying a critical gap in cancer therapy to building an innovative solution and business model that can reshape patient care.

For those who want to explore the details further, here is the full presentation featured in the video.

Pager

Great ideas deserve clarity. Our one-pager distills the essence of Oncoligo-our mission, solution, and market opportunity into a single page that captures both the science and the strategy behind our vision. See the full snapshot in the PDF below.

Development Road-map & Milestones

To guide our innovation from early-stage research to commercial availability, we’ve outlined a comprehensive development timeline. This roadmap defines critical milestones across all phases - design, validation, and spreading.

Before building our own roadmap, we first explored the core stages of drug development. Although many drugs exist today, the journey to develop a new one is long and complex, typically taking 10 to 15 years. It includes:

  • Drug Discovery - identifying a biological target and selecting promising candidate molecules.
  • Preclinical Development - testing safety, toxicity, and formulation in the lab and in animal models.
  • Clinical Development - evaluating the drug in people across three phases of clinical trials.
  • Regulatory Approval & Phase IV – submitting all data to authorities and monitoring long-term effects after approval.

While these phases sometimes overlap, they outline the essential path every new therapy must follow [28].

In the next diagram, we present a tailored version of this process, adapted to our project, with specific biological, clinical, and business milestones.

To better structure our development process, we divided our timeline into two main tracks.

The first focuses on the scientific aspects of our work, from biological experiments to computational modeling. This track outlines our key research and development efforts over the next five years.

2025-2026 - These years are focused on in vitro validation - starting from GFP models to more disease-relevant cancer cell lines. This is where our ASO pipeline is tested, refined, and adapted for real therapeutic targets.

2027-2028 - We will initiate in vivo experiments in mouse models, assessing efficacy and biodistribution. Simultaneously, we will expand our model to additional cancer types, tailoring the design pipeline to various tumor contexts. This is the transition point from bench to biology.

2028 - We’ll conduct toxicology and pharmacokinetics studies, including GLP-compliant protocols, to ensure safety and regulatory readiness. This is a critical milestone before engaging with the FDA.

2029-2030 - Focus shifts to formulation optimization: fine-tuning dosage, the chemistry of antibody–ASO conjugates, and delivery systems. These refinements are essential to ensure therapeutic stability, targeting specificity, and clinical feasibility.

In parallel to our scientific development, we’ve outlined a long-term plan supporting the translation of our Minimum Viable Product (MVP) - the MTAP-targeted ASO-antibody complex for lung cancer - into a clinically viable product.

This MVP represents the first complete implementation of our modular therapeutic platform, demonstrating both therapeutic efficacy and commercial feasibility, and serving as the foundation for expansion into additional cancer types.

We begin with market research, competitive landscape analysis, and defining a preliminary business model, weighing in-house development versus strategic licensing. In parallel, we initiate academic and clinical collaborations and secure early-stage IP through a provisional patent.

As we progress into 2026, we focus on fundraising and team expansion, preparing the operational base for long-term growth. By 2028, we initiate regulatory preparation and define our IP holding structure, leading to the Pre-IND submission to the FDA in support of planned preclinical studies.

Between 2029 and 2030, our efforts will center on preclinical development – including in vivo efficacy, toxicology, pharmacokinetics, and formulation optimization – to ensure safety and readiness for human trials.

Clinical development begins in 2030–2031 with Phase I studies to establish safety and dosing, followed by Phase II trials in 2032–2033 to evaluate efficacy, and Phase III trials in 2034–2035 to confirm therapeutic benefit at scale. Alongside these stages, we will engage with clinical-stage partners and health insurers to explore potential early access pathways.

Looking ahead to 2036–2037, we plan to submit a Biologics License Application (BLA). Upon regulatory approval, our therapy will be ready for market entry.

Commercial operations are expected to commence from 2038 onward, including large-scale manufacturing, distribution, marketing, and post-marketing surveillance. This marks the transition from development to patient access – ensuring our innovation reaches those who need it most, safely and sustainably.

Financial Plan

Behind every vision lies a solid foundation. This financial plan presents Oncoligo’s development costs, projected revenues, and funding roadmap -demonstrating how strong science meets strategic, scalable growth. Dive into the full breakdown in the PDF below.

Risks Analysis

Every innovation carries uncertainty. This document maps the scientific, regulatory, and financial risks that could influence Oncoligo’s path and the strategies we’ve built to overcome them. Explore how we turn challenges into opportunities for smarter, safer progress in the PDF below.

Regulatory & IP Strategy

The successful translation of Oncoligo from a pioneering iGEM project into a future clinical and commercial reality requires not only strong science but also a well-structured strategy for intellectual property and regulatory advancement. Intellectual property protection secures the unique innovations that differentiate our platform, while regulatory planning ensures a clear path from preclinical discovery to patient impact. Together, these pillars create the foundation for sustainability, competitiveness, and eventual real-world application.

In the following sections, we outline our comprehensive patent plan, covering the sequence level, epitope discoveries, antibody design, modular platform topology, and computational algorithms. We then present our roadmap through the FDA/EMA regulatory pathway, followed by a freedom-to-operate (FTO) assessment that highlights our ability to develop and commercialize Oncoligo without infringing existing patents.

Patent Plan

To ensure the long-term success and defensibility of Oncoligo, we have developed a structured patent strategy that spans every layer of our technology-from molecular sequences to computational design. This approach not only safeguards our therapeutic assets but also creates opportunities for licensing, strategic collaborations, and commercial growth.

Click below to view our full patent plan.

FDA / EMA Pathway

While our project is currently at the preclinical proof-of-concept stage, we have already considered the regulatory framework needed to translate our platform into a future clinical application. This section outlines the regulatory and operational roadmap for this process, divided into three key parts: Preclinical Requirements, and Clinical Trial Phases.

Preclinical Requirements

Before advancing to clinical studies, further in vivo experiments will be necessary to evaluate Oncoligo’s efficacy, biodistribution, safety, and toxicity in animal models. These studies are essential to establish pharmacokinetics (PK), pharmacodynamics (PD), and potential off-target effects.

Clinical Trial Phases

Our clinical roadmap is structured across four sequential phases, each with specific objectives that build upon one another. This progression is essential to first confirm safety and tolerability in patients, then evaluate efficacy and biological mechanisms of action, and finally validate outcomes at scale before transitioning into post-marketing surveillance. By following this stepwise pathway, we ensure that Oncoligo is developed responsibly and in alignment with FDA and EMA requirements.

Click below to expand and view the full clinical trial plan.

Freedom-to-Operate Summary

A strong freedom-to-operate (FTO) position is essential to ensure that Oncoligo can be developed and commercialized without infringing existing patents. By systematically reviewing prior filings, expired claims, and available space for new IP, we confirmed that our platform is positioned for growth, licensing, and eventual clinical translation.

Click below to expand and view the full FTO analysis.

People & Stakeholders

Our advisory board brings together leading experts in computational biology, antibody engineering, oncology, entrepreneurship, and innovation, guiding our team with decades of academic, clinical, and industry experience. Their specific contributions to our project are detailed on our Attributions page, where we elaborate on the unique guidance each mentor provided - from computational model design and antibody sequence optimization to clinical insights, entrepreneurial advice, and strategic project development.

Team Capabilities

Our team consists of 11 dedicated and talented students, ranging from B.Sc. to Ph.D. levels, bringing together diverse expertise from multiple fields. Each member contributes strong skills, creativity, and motivation to address the urgent health challenge of lung cancer and to drive forward the development of our therapeutic solution.

We possess a wide-ranging skill set that includes scientific research, experimental design, programming and modeling, bioengineering, and technical development, as well as knowledge in marketing strategies and outreach. This multidisciplinary approach is one of our greatest strengths, allowing us to tackle challenges from different perspectives.

Our team includes:

  • A Ph.D. student in Genetic Cell Engineering in Cancer, head of our wet lab team, with deep expertise in wet lab experimentation, experiment management, and data analysis.
  • A Master’s student in Mathematics, head of our modeling team, with a strong programming and computational background.
  • A Master’s student in Human Molecular Genetics and Biochemistry, with extensive wet lab experience.
  • A Master’s student in Computer Science, with strong computational and data-driven problem-solving skills.
  • Several Bachelor students in Biotechnology, Bioengineering, and Computer Science, bringing technical expertise, fresh ideas, and hands-on lab experience.

We are guided by our Principal Investigator, Prof. Tamir Tuller, a leading researcher in engineering, computer science, and synthetic biology. Prof. Tuller is an experienced entrepreneur and innovator, serving as co-founder and CSO of Synvaccine Ltd. and Imagindairy Ltd., as well as CTO of MNDL Bio.

While our team’s strengths lie in our multidisciplinary knowledge and collective passion, we also recognize areas for improvement. We have limited legal expertise in navigating regulatory frameworks and less experience in large-scale production. To address these challenges, we aim to strengthen our knowledge through targeted training and workshops, as well as by seeking partnerships with legal experts and industry professionals. These collaborations, along with our strong foundation, will help us bridge gaps and increase the efficiency of our work.

Looking ahead, we recognize that as Oncoligo moves from concept to implementation, new capabilities will be needed to complement our scientific and technical foundation. To transition from preclinical research toward future therapeutic development, additional expertise will be required in regulatory affairs (to navigate preclinical and clinical approval pathways), pharmacology (to optimize dosage, mechanism of action, and efficacy), and toxicology (to evaluate safety profiles and potential off-target effects).

Beyond these scientific needs, we also aim to strengthen our business and operational capacity. This includes specialists in intellectual property (to protect our innovations and guide patent strategy), fundraising and investment (to secure long-term financial sustainability), and project management (to coordinate collaborations, timelines, and resources as the project scales).

In the long term, we plan to expand our network with clinicians, biotech advisors, and industry partners who can provide practical insights into clinical translation, manufacturing standards, and patient accessibility. By strategically integrating these future recruits and collaborators, we will ensure that Oncoligo evolves from an academic initiative into a robust, translation-ready venture.

Social Impact

Oncoligo is more than a scientific or entrepreneurial project -it is a commitment to people and community. From the very beginning, we worked closely with patients and clinicians, listening to their voices and experiences. Their perspectives shaped our decisions, reminding us that the value of innovation lies not only in scientific progress but also in improving quality of life, accessibility, and hope for those facing cancer.

Our impact also extends to education and community building. ISRAGEM, Israel’s first iGEM-inspired national competition for high school students, has become a cornerstone of our outreach. In 2025, we proudly expanded this initiative, reaching over 250 students from more than 10 schools, with a special focus on engaging underrepresented groups from the social and economic periphery. Through interactive workshops, mentorship, and a final symposium at Tel Aviv University, ISRAGEM gave young students their first hands-on experience in synthetic biology and encouraged many to pursue careers in science and engineering. In addition, we are supporting the ORT school network in developing a new synthetic biology curriculum - advising teachers on content and meeting directly with students to foster early engagement with the field. By empowering diverse young minds, we aim not only to advance our own project, but also to build a stronger and more inclusive scientific community for the future.

In parallel, we embraced our role within the global iGEM community. We organized the Global Hybrid Mini-Jamboree at Tel Aviv University, hosting teams from across the world online and welcoming the Technion team in person. This event fostered dialogue, collaboration, and scientific exchange across borders. Following the iGEM competition, we plan to publish articles about our project in the press, ensuring that our message reaches the wider public beyond academia. We also participated in multiple international conferences, ensuring that our work contributes to the broader conversation on RNA therapeutics and synthetic biology.

Taken together, these efforts highlight that Oncoligo is not just about developing a therapeutic platform. It is about responsibility, inclusivity, and community. Our project strives to educate, connect, and inspire-showing that synthetic biology can be a force for positive change far beyond the lab. For more details, see our Human Practices section.

To conclude, we chose to capture our social impact in a single illustration. This graphic brings together the four pillars that guide our work-patients and clinicians, education, community, and outreach, showing how Oncoligo extends beyond the lab to create lasting connections with people and society.

Long Term Impacts

The long-term vision of Oncoligo extends far beyond a single therapeutic program. By combining synthetic lethality targeting, the BROTHER safety switch, antibody-guided delivery, immune-epitope activation, and a unique computational design model into one integrated platform, we have established a modular therapeutic architecture with broad adaptability.

In oncology, our approach can be rapidly expanded from lung cancer into additional high-burden cancers such as colorectal, pancreatic, and breast cancer, where tumor suppressor loss and rare mutations leave patients with few treatment options. Because the platform acts on shared synthetic lethal partners rather than the mutations themselves, it can cover a wide spectrum of patient subgroups, reducing the fragmentation that limits current targeted therapies.

To illustrate this potential expansion, we analyzed patient data from The Cancer Genome Atlas (TCGA) - a global initiative that maps the genetic landscape of thousands of tumors across diverse cancer types. This analysis allowed us to estimate, for each cancer type, the proportion of patients carrying loss-of-function mutations in tumor suppressor genes with at least one known synthetic lethal partner, as well as the estimated annual number of patients worldwide that are potentially relevant to our platform (shown above each bar). The figure below presents a selected subset of major cancer types for clarity, although additional types are also relevant to our approach.

Figure 11: Estimated percentage and annual number of patients potentially relevant to Oncoligo’s platform across major cancer types. Bars represent the fraction of patients carrying loss-of-function mutations in tumor suppressor genes that have at least one known synthetic lethal partner, with the corresponding estimated annual number of relevant patients worldwide shown above each bar, based on TCGA and GLOBOCAN data. For clarity, the figure presents a selected subset of major cancer types, while additional types are also relevant to our approach.

Beyond oncology, the modularity of our computational design and antibody-oligonucleotide conjugation system allows expansion into genetic diseases where selective RNA silencing is required. To assess the potential global relevance of such applications, we relied on large-scale epidemiological analyses from the European Journal of Human Genetics [29]. These studies report that rare diseases collectively affect 3.5–5.9% of the global population, corresponding to approximately 280–480 million individuals worldwide. Recent analyses suggest that around 10–15% of pathogenic variants are potentially amenable to antisense oligonucleotide (ASO) therapy, primarily through splice-switching or RNA-silencing mechanisms [30].
Combining these estimates provides the following calculation:
8.1 billion people×4.7% (average prevalence) ×12% (share of ASO-amenable disorders) ≈ 45 million individuals.

Therefore, in addition to millions of oncology patients, approximately 40–50 million people worldwide could, in principle, benefit from ASO-based interventions targeting genetic diseases.

Importantly, our ASO design engine is not limited to cancer or even to human medicine: it can support any researcher or developer working with antisense oligonucleotides across organisms, from human cells to yeast, for therapeutic, industrial, or basic science purposes. Looking further ahead, the principles and models we have developed may enable future applications in related modalities such as siRNA, dsRNA, or even gRNA, broadening the reach of computational nucleic acid design.

To better understand the potential scope of researchers and developers who could benefit from such a platform, we examined the global landscape of RNA therapeutics research and innovation, Over the past 15 years, more than 25,000 peer-reviewed publications on antisense oligonucleotides and RNA therapeutics have been indexed in PubMed, reflecting a rapidly expanding global research community. According to Scopus and Nature Index data, over 300 universities and research institutes are actively publishing in this field, alongside approximately 150 biotechnology companies developing RNA-based therapeutics and experimental tools.

Together, these insights highlight the broad scientific ecosystem that could benefit from accessible, data-driven platforms like Oncoligo, fostering faster discovery, improved reproducibility, and cross-disciplinary collaboration in RNA research.

The scope of our platform can be best illustrated through its expansion path. Starting with lung cancer as the entry point, Oncoligo progressively extends into other cancers, genetic diseases, and ultimately positions itself as a universal RNA design engine. The figure below summarizes this trajectory and highlights the adaptability and scalability of the platform.

Figure 12: Expansion Path of Oncoligo. The platform begins with lung cancer, expands to other cancers with synthetic lethal partners, diversifies into genetic diseases, and evolves into a universal RNA design tool that supports applications across medicine, biotechnology, and different organisms.

If successful, the long-term global impact will be substantial: raising survival rates worldwide, decreasing the economic burden of late-stage cancer care, and making precision medicine faster, safer, and more cost-effective. Oncoligo’s vision is not only to bring a new therapy to patients, but to establish an enduring platform that reshapes the way we fight cancer and genetic disease, and more broadly, the way we design and apply RNA-targeting tools for decades to come.