At ONCOLIGO, our TAU iGEM 2025 team, we believe that science doesn't exist in a vacuum. Throughout the design and development of our therapy for lung cancer, we continuously asked ourselves: Is our project relevant, responsible and good for the world? To address this question, we actively engaged with the broader scientific, clinical, educational, and patient communities. We sought diverse perspectives from oncologists, pharmacologists, drug developers, biotech companies, and patients themselves. These conversations shaped critical decisions in our project - from choosing the cancer type we target to how we evaluate delivery strategies and plan our experiments. Our Human Practices efforts also extended beyond our own project. We organized educational activities to promote synthetic biology, collaborated with international iGEM teams, and helped support the next generation of researchers in Israel. As part of this commitment, we hosted the iGEM Global Hybrid Mini-Jamboree Meetup at Tel Aviv University. The event brought together more than 10 international teams online alongside the Technion iGEM team in person for a day of scientific exchange, peer feedback, and community building. We also prepared and shared a practical guide for hosting hybrid meetups, ensuring that future iGEM teams can benefit from our experience and strengthen the collaborative spirit of the competition. On this page, we document how ethical considerations, stakeholder feedback, and public engagement influenced our project design and purpose, and how we aim to ensure our work benefits society in a meaningful and responsible way.
Throughout the season, we consulted with a wide range of stakeholders, clinicians, academic experts, biotech professionals, and patients – to ensure that our project is scientifically robust, clinically relevant, and socially responsible. Their feedback directly shaped key decisions in our design, from choosing lung cancer as our focus indication to refining delivery strategies and considering patient quality of life.
We engaged with leading oncologists and clinicians who helped us align our project with real patient needs. Their input guided us in selecting the cancer type to focus on and in refining our therapeutic goals.
Emphasized patient-centered treatment strategies in lung cancer.
Prof. Amir Onn, Head of the Institute of Pulmonology at Sheba Medical Center and an expert in pulmonary oncology, met with our team on several occasions to discuss our project and offer invaluable clinical insights. Drawing from his extensive experience treating lung cancer patients, he introduced us to the most common mutations observed in practice, how mutations evolve throughout disease progression, and the importance of tailoring treatment lines for each patient. His personalized medicine approach deepened our understanding of lung cancer biology and guided us in framing our therapeutic strategy. Additionally, Prof. Onn delivered an educational session on lung cancer management from the physician’s perspective, helping us align our work with real clinical needs.
Provided a broader oncology perspective and directed us to critical data resources.
Prof. Ido Wolf, Head of the Oncology Division at Tel Aviv Sourasky Medical Center, was introduced to our project and offered valuable guidance during its early stages. He helped us better understand the clinical landscape of oncology and directed us to key cancer databases and resources that supported our research. His input contributed to shaping our initial exploration of relevant cancer types and therapeutic opportunities.
Highlighted blood–brain barrier challenges, leading us to pivot away from brain tumors.
To explore the feasibility of targeting brain tumors with antisense oligonucleotides (ASOs), we consulted with Prof. Stuart Grossman, a leading neuro-oncologist at the Johns Hopkins Kimmel Cancer Center. Prof. Grossman shared his experience from a Phase II clinical trial involving Aprinocarsen, an ASO targeting PKCα, which showed no clinical benefit when administered intravenously to patients with brain tumors. He highlighted the key challenge of blood-brain barrier (BBB) penetration and explained that current delivery routes, such as intrathecal injection have major limitations in effectively reaching widely distributed tumors like glioblastoma (GBM), without compromising BBB integrity.
Based on his input, we reassessed our approach. Since our project’s primary focus is on ASO design and optimization, rather than drug delivery systems, we concluded that it would be more impactful and responsible to target cancers with simpler and more accessible delivery requirements.
Academic researchers in pharmacology, antibody engineering, and RNA therapeutics provided expertise that strengthened our molecular design and experimental plan.
Advised on antibody engineering and provided the initial plasmid sequence.
Prof. Itai Benhar, an expert in antibody engineering at Tel Aviv University, delivered a lecture to our team about his research on bispecific antibodies and antibody-drug conjugates. He provided critical guidance on the design of the antibody component in our therapeutic complex and shared the initial plasmid sequence we used as a basis for our construct. His expertise played a key role in shaping the molecular design of our targeting strategy.
Introduced us to the Wnt/APC pathway and shared resources to explore synthetic lethality in colorectal cancer, expanding the potential scope of our platform.
Prof. Rina Rosin-Arbesfeld, a colorectal cancer expert at Tel Aviv University, met with our team to discuss potential applications of our ASO approach in colon cancer. She shared insights from her research on the Wnt signaling pathway, particularly the APC gene, and introduced us to relevant cell lines, reagents, and delivery methods used in her lab. Prof. Rosin-Arbesfeld also expressed interest in helping us test synthetic lethality partners of APC in her ongoing studies.
Prof. Jack S. Cohen, a pioneer in antisense pharmacology, shared historical and mechanistic insights that helped us contextualize ASO development timelines, common pitfalls, and realistic translational milestones.
On September 4th, we met with Prof. Jack S. Cohen, an expert in pharmacology and antisense oligonucleotides. Prof. Cohen earned his doctorate at Cambridge University and held research and faculty positions at the NIH, Harvard Medical School, Georgetown University, and later in Israel at Sheba Medical Center, Hebrew University, and Ben-Gurion University. With over 200 publications and a long history working on antisense oligonucleotides, he shared with us his unique perspective on the discovery and development of ASOs.
Contributed guidance on target selection and pharmacokinetic considerations, linking our scientific ideas with practical drug development strategies.
Pharmacology doctor at Ha'Ivrit University, Jerusalem. former CEO of LifeWave and current head of Drug Development Department at Starget Pharma. Advised about Target search and provided Pharmacokinetic input.
Recommended antibody-ASO conjugation as the most clinically relevant delivery approach.
We had the opportunity to meet Dr. Oren Bogin, former CEO of Immunorizon and an expert in protein chemistry and biologics development. With his extensive experience in drug discovery and biological therapeutics, Dr. Bogin provided valuable advice on the delivery challenges of ASO-based therapies and reviewed the various delivery technologies currently available in the market. He strongly encouraged us to pursue an antibody-conjugation strategy, which aligns with our modular platform approach, as it offers both targeted delivery and clinical relevance. His insights helped refine our delivery strategy and strengthened the rationale behind integrating an antibody into our therapeutic complex.
Shared insights on RNA therapy protocols and translational considerations.
We had the privilege of consulting with Dr. Nofar Mor from Sheba Medical Center, who specializes in developing RNA-based therapies for ultra-rare diseases. Dr. Mor generously shared her expertise on antisense oligonucleotide (ASO) protocols, helping us refine our experimental design and better understand the practical aspects of RNA therapeutics. In addition to her clinical work, Dr. Mor is a co-author of the book Target Identification and Validation in Drug Discovery: Methods and Protocols [1], which provided us with valuable insights into modern approaches for drug discovery and validation. Her guidance significantly contributed to shaping our project’s therapeutic strategy and strengthened the translational relevance of our work.
Industry partners gave us practical advice on feasibility, scalability, and delivery technologies.
Supported us with ASO design protocols and experimental controls.
We collaborated with SKIP Therapeutics, a biotech company developing RNA-based therapies using antisense oligonucleotides (ASOs). The SKIP team supported us with expert guidance on ASO design and experimental execution in mammalian cell lines. They provided detailed protocols and generously supplied us with a MALAT1-targeting ASO as a positive control and a scrambled ASO as a negative control. Their support significantly enhanced the reliability and robustness of our in vitro experiments.
Introduced us to strategies for antibody–oligonucleotide conjugation and their trade-offs.
As part of our entrepreneurship and technology exploration, we held discussions with Creative Biolabs, a leading biotech company specializing in Antibody-Oligonucleotide Conjugates (AOCs). AOCs represent an emerging platform that combines the precision of oligonucleotides with the targeted delivery capabilities of antibodies, creating a powerful tool for therapeutic applications.
During these conversations, we learned about the different strategies for AOC synthesis-including ion-interaction coupling, avidin–biotin bridges, direct conjugation to amino acids, and hybridization-based assembly. Each method has unique advantages and trade-offs in terms of stability, cost, and lysosomal release efficiency.
These insights helped us evaluate how AOCs could complement our ASO-based therapeutic platform, particularly in enhancing delivery, improving tissue specificity, and ensuring therapeutic stability in vivo. Importantly, we discussed their potential applications in oncology, from gene silencing and targeted ASO delivery to immune modulation and biomarker detection.
By engaging with industry experts such as Creative Biolabs, we not only gained a clearer understanding of the practical challenges and opportunities in bringing AOCs into the clinic, but also strengthened the entrepreneurial perspective of our project - bridging the gap between academic innovation and translational biotechnology.
Speaking directly with patients helped us connect the science to the human experience of cancer.
Ellen Nemetz, a lung cancer patient with a rare KRAS mutation, emphasized the urgent need for therapies with lower toxicity and broader applicability across mutation types. Her story motivated us to design for reduced side effects and for inclusion of both common and rare variants.
As part of our Human Practices work, we interviewed Ellen Nemetz, a 64-year-old artist from Arizona, USA, who was diagnosed with stage IV lung cancer 1.5 years ago. Ellen is a never-smoker and carries the KRAS-Q61H mutation, a rare and challenging mutation for which treatment options remain limited.
Ellen first experienced Bamberger–Marie syndrome, a rare condition affecting about 5% of lung cancer patients, causing swelling and pain in the legs. Initially, her doctors investigated her heart, liver, and kidneys, all of which appeared normal. When she also mentioned a persistent dry cough, imaging revealed a primary carcinoma.
Initially staged as 3B, a subsequent PET scan revealed a metastasis in her hip, upgrading the diagnosis to stage IV. At that time, the disease had not yet spread to her brain or major organs.
Ellen began treatment with a combination of cisplatin, Pemetrexed, and Keytruda (immunotherapy).
Ellen shared that cisplatin (chemotherapy) was the most difficult chemotherapy, with side effects sometimes compounded by anti-nausea medications. Constipation and fatigue were especially challenging. She emphasized the importance of staying active - returning to the gym helped her both physically and mentally.
Ellen emphasized the need for new treatments that address rare mutations like KRAS-Q61H, as current options remain limited.
She hopes future research will consider patients’ quality of life, provide clear communication about risks and benefits, and make innovative therapies accessible for rare mutation carriers.
Ellen Nemetz, provided explicit written consent to be identified by name and to include limited details above. All other participants remain anonymous.
In addition to Ellen’s story, we interviewed another lung cancer patient (“S”), diagnosed with an EGFR-mutant tumor. She described the contrast between targeted therapy, which initially restored her quality of life, and chemotherapy, which brought severe fatigue and functional decline. Beyond treatment efficacy, she highlighted the heavy toxicities, financial burden, and bureaucratic challenges patients face. Her perspective reinforced our focus on designing therapies that are not only effective but also less toxic, more accessible, and adaptable across mutation types.
We also interviewed a lung cancer patient who preferred to remain anonymous (“S”). She was diagnosed two and a half years ago after months of unexplained symptoms and multiple hospitalizations. The diagnosis came abruptly and insensitively, following an MRI that revealed a large stage IV lung tumor.
Initially, S responded remarkably well to a targeted therapy (dacomitinib), with tumors shrinking by more than 60% within days. However, after a year the disease progressed, with new metastases and multiple biopsies required to confirm recurrence. Because her tumor carried an EGFR mutation that is often resistant to immunotherapy, she faced difficult treatment decisions. Although offered a place in an early-stage clinical trial, she declined due to the uncertainty and instead began chemotherapy. She described this as “a completely different world”: compared to targeted therapy, chemotherapy brought severe fatigue, cognitive fog, and daily functional limitations.
Despite these challenges, S has managed to continue working part-time and relies heavily on supportive activities such as water exercise to maintain her well-being. She emphasized the heavy bureaucratic and financial burden of treatment, including prolonged struggles with insurance coverage and drug access. At times she felt that the responsibility for coordinating her care fell primarily on her and her sister, rather than the health system.
Her message to researchers developing new therapies was clear: beyond efficacy, it is critical to reduce toxicity, simplify access, and design solutions that are realistic for patients managing complex, long-term disease.
We integrated insights from clinicians, patients, and industry to iteratively refine ONCOLIGO. Below we document concrete design decisions that changed because of Human Practices:
We interviewed clinicians, industry scientists, pharmacologists, and patients. All participants gave informed consent; we collected no identifiable medical data. Notes were anonymized and double-checked by two team members to reduce bias. We follow iGEM’s No Human Experimentation and Human Subjects Research policies.
Alongside our core values – saving lives, deep scientific understanding, and safe and responsible work – we believe it is essential to remain connected to the community. With this vision in mind, we founded ISRAGEM, the first iGEM-inspired national competition in Israel for high school students, with a special focus on engaging underrepresented groups from the social and economic periphery. Working together with the Tel Aviv University Commission for Equality, we aimed to ensure diversity among future generations of science students.
At the same time, we see being part of the iGEM community as a privilege and an opportunity to learn from and collaborate with teams worldwide. Therefore, we organized the iGEM Global Hybrid Mini-Jamboree Meetup at Tel Aviv University, hosting over 10 teams from around the world online and welcoming the Technion iGEM team in person. This event strengthened scientific exchange, feedback, and community building across borders.
At iGEM TAU, we believe science should be inclusive, inspiring, and accessible to everyone - regardless of background. With this vision in mind, we founded ISRAGEM, the first national synthetic biology competition for high school students in Israel.
ISRAGEM (Israeli Synthetic Biology Challenge) is an educational outreach initiative designed and led by the iGEM TAU team. It introduces high school students to the world of synthetic biology through interactive workshops, mentorship, and a student-led project competition. The program ends with a final event where participants present their synthetic biology ideas to a panel of scientists, educators, and industry professionals.
Israel’s diverse society is rich in potential, but many students - particularly those from underrepresented communities - lack exposure to advanced scientific fields like synthetic biology. ISRAGEM aims to bridge that gap by creating a shared educational space where young minds can collaborate, innovate, and engage with cutting-edge science.
By lowering the barriers to entry and providing mentorship, we hope to cultivate a new generation of synthetic biologists who will shape the future of biotechnology in Israel and beyond.
Our goal is to make ISRAGEM a sustainable, national initiative that continues annually. We are working with educational institutions and government bodies to scale the program, develop translated materials, and offer teacher training to expand its reach and long-term impact.
[1] J. Moll and S. Carotta, Eds., “Target Identification and Validation in Drug Discovery,” vol. 2905, 2025, doi: 10.1007/978-1-0716-4418-8.