What is Theranostics?
Theranostics is a new subspecialty within oncology and nuclear medicine that collides the power of molecular imaging to detect tumors with the power of radionuclides to attack cancer cells, using the same molecular platform.
Theranostics applications
Currently approved applications
β emitters
Theranostics: Core Concept
Theranostics uses radio-labeled molecules that:
-
Bind specific tumor markers (diagnostic imaging).
-
Deliver cytotoxic radiation directly to tumor cells (therapy).
These agents generally pair a diagnostic radionuclide (like Gallium-68) with a therapeutic radionuclide (like Lutetium-177), targeting specific receptors overexpressed in certain cancers.
Theranostic agents such as Lutathera® and Pluvicto™ represent major clinical advances by allowing targeted treatment and real-time tracking of tumor response.
GEP-NETs
1. Lutathera® (Lutetium-177-DOTATATE)
Indication: Neuroendocrine tumors (NETs) — especially gastroenteropancreatic NETs (GEP-NETs).
Mechanism:
• Targets somatostatin receptor subtype 2 (SSTR2), highly expressed in NETs.
• Lutathera is a radiolabeled somatostatin analog: DOTATATE conjugated with Lutetium-177, a β-emitter.
• Diagnostic partner: Ga-68-DOTATATE PET/CT scan (for selecting SSTR-positive patients).
Theranostic Workflow:
1. Imaging with Ga-68-DOTATATE PET to identify SSTR-expressing tumors.
2. If positive, treat with Lu-177-DOTATATE to deliver targeted radiation.
Clinical Benefits:
• Improved progression-free survival (PFS) and overall survival (OS).
• Lower toxicity than systemic chemotherapy.
• Beneficial in well-differentiated, inoperable or metastatic NETs.
_______________________________________
mCRPC
⚕️ 2. Pluvicto™ (Lutetium-177-PSMA-617)
Indication: Metastatic castration-resistant prostate cancer (mCRPC).
Mechanism:
• Targets prostate-specific membrane antigen (PSMA), overexpressed in 85–90% of prostate cancers.
• Pluvicto combines PSMA-617 ligand with Lutetium-177.
• Diagnostic partner: Ga-68-PSMA-11 PET/CT scan or F-18-DCFPyL PET.
Theranostic Workflow:
1. PSMA PET imaging to confirm PSMA expression in tumors.
2. Treat with Lu-177-PSMA-617 in eligible patients.
Clinical Benefits:
• Shown to significantly improve survival and delay disease progression in heavily pretreated mCRPC patients.
• Favorable safety profile compared to conventional chemotherapy.
• Approved by FDA in 2022 (based on the VISION trial)
The future of theranostics lies in more potent, precise, and personalized approaches. Alpha emitters promise higher efficacy, while combination therapies may overcome resistance mechanisms. As research expands into diverse tumor types, theranostics is set to play a pivotal role in the next generation of precision oncology.
Upcoming novel applications
α emitters
Future Directions in Theranostics
1. Alpha-Emitting Isotopes (example: Actinium-225) for More Potent Localized Radiation Alpha emitters like Actinium-225, Bismuth-213, and Astatine-211 are gaining momentum in theranostics due to their high linear energy transfer (LET) and short tissue penetration range (~50–100 µm). This allows them to deliver highly cytotoxic radiation specifically to cancer cells, sparing surrounding healthy tissues.
Advantages:
High potency: Alpha particles cause double-stranded DNA breaks, which are more difficult for cancer cells to repair.
Minimal collateral damage: Due to limited travel distance, alpha radiation reduces damage to nearby normal cells.
Applications under study:
-
Prostate cancer (example: targeted alpha therapy using Actinium-225–labeled PSMA ligands).
-
Leukemias and lymphomas, due to the ability of alpha particles to eradicate minimal residual disease.
-
Combination with nanocarriers to improve delivery specificity and reduce systemic toxicity.
2. Combination Therapies with Immunotherapy or PARP Inhibitors
The synergy between theranostics and other targeted therapies is a promising area of development.
Theranostics + Immunotherapy:
-
Radiation from theranostic agents can upregulate neoantigen presentation, enhancing immune system recognition.
-
Radiation-induced immunogenic cell death may boost the efficacy of checkpoint inhibitors (example: PD-1, CTLA-4).
Trials are ongoing combining radionuclide therapy with CAR-T cells or immune checkpoint blockade in solid tumors.
Theranostics + PARP Inhibitors:
-
PARP inhibitors (example: olaparib) impair DNA repair in tumors with BRCA mutations or homologous recombination deficiencies.
-
Combining with radionuclide therapy exploits synthetic lethality, overwhelming the cancer cell's ability to repair radiation-induced DNA damage.
-
Especially relevant in ovarian, breast, and prostate cancers.
3. Theranostics Under Development for New Tumor Types
While prostate and neuroendocrine tumors have seen the most clinical success so far, research is expanding into more challenging cancers:
a. Thyroid Cancer
Traditionally treated with radioactive iodine (I-131), now being re-explored with:
Next-gen iodine isotopes (I-124 for PET imaging).
Improved tracers for dedifferentiated thyroid cancers that lose iodine avidity.
Sodium-iodide symporter (NIS) gene therapy combined with radioiodine.
b. Breast Cancer
Development of HER2-targeted radioligands, enabling diagnosis and therapy in HER2-positive cases.
Investigational use of FAP-targeted theranostics (fibroblast activation protein), relevant for triple-negative breast cancer.
Imaging agents using 89Zr-labeled trastuzumab to map HER2 expression.
c. DLL3-Expressing Cancers
Delta-like ligand 3 (DLL3) is an inhibitory Notch ligand aberrantly overexpressed in several aggressive cancers, particularly small cell lung cancer (SCLC) and other neuroendocrine tumors, while having minimal expression in normal tissues. Its tumor-specific expression makes it an attractive target for novel radionuclide-based theranostics.
d. Glioblastoma
One of the most aggressive and therapy-resistant brain tumors.
Challenges: blood–brain barrier (BBB), heterogeneity.
Emerging approaches:
-
Theranostic nanoparticles that cross the BBB.
-
Targeted alpha therapy with ligands like substance P or IL-13 conjugates.
-
Research into EGFRvIII or integrin αvβ3 targeting for glioblastoma imaging and treatment.
e. Newly emerging targets and radio isotopes
-
GRPR
-
FAP
-
Integrin
-
etc.
