What’s Next in Cancer Care? Key Oncology Advances to Watch in 2026

The start of a new year invites assessment of trends likely to shape clinical oncology. Based on developments seen in 2025, the following ten advances are the most probable influences on practice in 2026. These perspectives reflect converging signals from clinical trials, regulatory activity, technology and real-world practice rather than speculation.

1. Artificial intelligence will become a meaningful clinical support for patients and oncologists.
AI assistants are likely to be widely integrated as decision‑support and patient‑navigation tools, especially where specialist access is limited. They will help explain diagnoses, treatment options, adverse events and prognosis in patients’ native languages and aligned with local care pathways, reducing barriers to timely care. Early AI guidance on managing common toxicities could lower unnecessary treatment interruptions and delays. Continuous learning from hundreds of thousands of oncology queries, integration with validated guidelines and RAG frameworks will improve performance. AI will augment but not replace oncologists: human judgment, ethics and empathy will remain central, while high‑quality clinician‑generated data will be essential for model validation.

2. Molecular decision‑making after surgery or radiation therapy will enter routine practice.
Circulating tumor DNA (ctDNA) as a marker of minimal residual disease is moving from investigational use to an actionable clinical tool. Randomized studies in colorectal and urothelial cancers have shown ctDNA can identify patients at high risk of recurrence and spare ctDNA‑negative patients from unnecessary adjuvant therapy. Emerging evidence for renal cell carcinoma and HPV‑associated oropharyngeal cancer suggests broader applicability. By 2026, molecularly guided escalation or de‑escalation of adjuvant treatment is likely to be standard in selected tumor types.

3. Expansion of theranostics and radioligand therapies.
New PET tracers are expected to broaden diagnostic applications and enable early systemic‑therapy response assessment across tumor types. Radioligand therapies are poised for wider adoption with anticipated new indications in metastatic prostate cancer, neuroendocrine tumors, breast, thyroid and gastrointestinal malignancies. Trials of additional isotopes such as actinium, yttrium and rhenium may expand therapeutic options beyond current agents.

4. The future belongs to deeply personalized therapy.
Regulatory approvals in 2025 continued a shift toward biomarker‑driven strategies, companion diagnostics and tumor‑agnostic approaches. Of 39 original solid‑tumor drug approvals tracked this year, roughly half were personalized therapies. Truly non‑personalized approvals were rare. Therapeutic decisions are increasingly driven by molecular profiles—mutations, amplifications, receptor expression and microenvironmental signatures—making personalization a central principle of oncology development despite ongoing challenges in testing access and interpretation.

5. Continued rapid growth of antibody‑drug conjugates.
Antibody‑drug conjugates (ADCs) remain a fast‑growing sector, reflected by a surge in clinical activity: hundreds of ADC trials were launched in 2024–2025, including more than 100 early‑phase studies presented at major meetings. Development focuses on new antibody scaffolds, more stable linkers and diversified payloads. Increased regulatory submissions, approvals and indication expansions—particularly into earlier disease settings and combinations—are likely in 2026.

6. Cellular technologies will advance but are unlikely to deliver approvals for solid tumors in 2026.
CAR‑T therapies continue to transform hematologic malignancies but face fundamental barriers in solid tumors, including tumor penetration and the immunosuppressive microenvironment. Early‑stage trials show activity but not yet transformative results. Therapeutic mRNA vaccines have shown promising immune responses and tolerability, with several programs in Phase 3; registration‑enabling data are more likely in 2027–2029. TIL and TCR approaches are finding niches and may produce unexpected approvals, but large‑scale impact in solid tumors remains a longer‑term prospect.

7. The search for new immunotherapy options will continue, including bispecific antibodies.
Beyond PD‑1/PD‑L1 and CTLA‑4, many novel checkpoint targets have produced mixed results. Established immunotherapies will continue migrating into earlier settings, and complex combination regimens are under active study. Bispecific antibodies that redirect T cells to tumor cells represent a promising frontier; agents such as tarlatamab for small‑cell lung cancer and several bispecifics in hematologic malignancies illustrate the approach. Additional bispecific approvals in solid tumors are plausible by 2026 given ongoing Phase 3 programs. Targeting the immunosuppressive tumor microenvironment also remains active, with recent accelerated approvals and late‑stage trials for agents modulating tumor‑associated macrophages and related pathways.

8. The first PROTAC degrader approval is likely.
PROTACs, which induce targeted protein degradation via the ubiquitin‑proteasome system, advanced in 2025. The estrogen‑receptor degrader vepdegestant met its Phase 3 endpoint in the VERITAC‑2 trial and was submitted for regulatory review. Multiple other degraders are in Phase 2. Approval of the first PROTAC in 2026 is a realistic possibility.

9. Radiation therapy will focus on personalization, precision and intensity across stages.
Stereotactic ablative radiotherapy (SABR/SBRT) has shown low high‑grade toxicity and excellent local control and will see expanding indications, including oligometastatic and select metastatic settings. Numerous positive prospective studies reported in 2025 support broader use and guideline incorporation. Attention is shifting from seeking abscopal effects to understanding treatment‑induced progression (“badscopal” effects), with research into radiation‑induced factors such as amphiregulin that may promote immune suppression and distant progression. Evidence suggests concurrent chemoimmunoradiation may blunt immunotherapy benefits; a move toward sequential strategies—induction chemoimmunotherapy followed by definitive radiation—appears likely to predominate for many locally advanced tumors.

10. Renewed, evidence‑driven interest in proton therapy.
Proton therapy experienced renewed attention as multiple prospective studies and over 900 trials—often in combination with immunotherapy—are under way. To date, comparative evidence versus modern photon techniques is inconsistent, and clear clinical superiority has not been established in many settings. Results expected in 2026 will be important to define the role and value of proton therapy.

These trends indicate an oncology landscape increasingly driven by precision, multimodal integration and technology‑enabled care, while preserving the central roles of clinical judgment, multidisciplinary teams and patient‑centered decision making.

Ilya Tsimafeyeu, Medical Oncologist, Member, Editorial Board, OncoDaily; Director, Bureau for Cancer Research – BUCARE, USA/Russia

Natalia Dengina, Radiation Oncologist, Editor‑in‑Chief, RUSSCO Newspaper; Head, Department of Radiation Therapy, Ulyanovsk Regional Cancer Center, Russia

Comments

Leave a Reply

Your email address will not be published. Required fields are marked *