4.1. Tumor-Agnostic Therapies
Tumor-agnostic (tumor of origin-agnostic) therapies—also known as tissue-agnostic or site-agnostic treatments—are a groundbreaking class of cancer therapies approved based on specific molecular biomarkers rather than the anatomical origin of the tumor. This precision-medicine strategy allows treatments to be matched to genetic alterations such as mutations, fusions, or immune signatures, enabling highly personalized therapy across diverse cancer types.
The first FDA approval of this kind was pembrolizumab (Keytruda) in 2017, granted for any unresectable or metastatic solid tumor displaying microsatellite instability-high (MSI-H) or deficient mismatch repair (dMMR)—marking a pivotal shift from tumor-site to biomarker-driven oncology [
78].
Since then, additional agents have secured tumor-agnostic FDA indications, including targeted therapies for NTRK gene fusions, BRAF V600E mutations, RET fusions, as well as treatments based on high tumor mutational burden (TMB-H) or HER2 overexpression (
Table 2) [
79,
80].
4.2. Lung Cancer
Lung cancer remains the leading cause of cancer-related mortality worldwide, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of cases and small cell lung cancer (SCLC) representing the remaining 15% [
81]. The advent of targeted therapies and immunotherapies has dramatically transformed treatment paradigms in both groups, providing options that are more precise and effective compared with traditional chemotherapy. Molecular profiling is standard of care in NSCLC, as multiple oncogenic drivers and immune biomarkers guide the selection of targeted and immune checkpoint therapies. Targeted therapies in lung cancer can be broadly categorized into (1) kinase inhibitors directed against specific oncogenic drivers (EGFR, ALK, ROS1, RET, MET, BRAF, KRAS, HER2, NTRK); (2) anti-angiogenic therapies targeting VEGF/VEGFR; and (3) immune checkpoint inhibitors acting on PD-1, PD-L1, and CTLA-4 pathways [
82,
83,
84,
85]. These categories reflect a shift toward personalized treatment selection, informed by comprehensive molecular testing, which is now a standard component of lung cancer management (
Table 3).
In NSCLC, one of the most important therapeutic breakthroughs has been the development of tyrosine kinase inhibitors (TKIs) directed against activating mutations in the
epidermal growth factor receptor (EGFR) gene [
82]. First-generation TKIs such as erlotinib and gefitinib demonstrated initial efficacy, followed by second-generation inhibitors like afatinib and dacomitinib, and ultimately third-generation osimertinib, which also overcomes the common resistance mutation T790M. Parallel advances have occurred in patients with gene rearrangements such as
ALK and
ROS1, where inhibitors including crizotinib, ceritinib, alectinib, brigatinib, lorlatinib, and entrectinib have achieved unprecedented clinical benefits. More recently, therapies targeting
RET fusions (selpercatinib, pralsetinib) and
MET exon 14 skipping mutations (capmatinib, tepotinib, savolitinib) have expanded the treatment arsenal [
86,
87,
88].
Another clinically relevant category involves BRAF V600E mutations, for which combined inhibition with dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor) has become the standard of care [
54]. Similarly, the recognition of
KRAS G12C mutations as actionable targets has led to the approval of novel covalent inhibitors, such as sotorasib and adagrasib, offering new options for a historically undruggable oncogene [
51,
89]. Additional therapeutic directions include HER2-directed therapies in
ERBB2-mutant NSCLC (trastuzumab deruxtecan, poziotinib in development) and NTRK inhibitors (entrectinib, larotrectinib) for patients with rare fusions [
57]. Angiogenesis inhibition remains a key therapeutic pillar, particularly in advanced NSCLC. Bevacizumab, ramucirumab, and nintedanib act by blocking the vascular endothelial growth factor (VEGF) axis, limiting tumor vascularization and enhancing the efficacy of chemotherapy and immunotherapy backbones [
84,
90]. Equally transformative has been the rise of immune checkpoint inhibitors (ICIs), which have dramatically reshaped the treatment landscape for both NSCLC and SCLC. Monoclonal antibodies against PD-1 (nivolumab, pembrolizumab) and PD-L1 (atezolizumab, durvalumab, avelumab) have demonstrated durable responses and survival benefits, particularly in tumors with high PD-L1 expression, high tumor mutational burden (TMB), or microsatellite instability (MSI) [
85]. In addition, the CTLA-4 inhibitor ipilimumab, alone or in combination with PD-1 blockade, has shown clinical activity, providing further immunotherapeutic strategies [
91].
In contrast, treatment options for small cell lung cancer (SCLC) remain more limited, reflecting the aggressive biology and paucity of actionable driver mutations. Nevertheless, immunotherapy has emerged as a meaningful advance: the addition of PD-L1 inhibitors such as atezolizumab or durvalumab to first-line chemotherapy has improved outcomes and set a new standard of care. Emerging research exploring DLL3-targeted therapies (e.g., tarlatamab) has shown promising results in second-line treatment for ES-SCLC, with response rates up to 40% and a median overall survival of 14.3 months [
92].
SCLC is characterized by rapid growth, early metastasis, and a high rate of initial response to chemotherapy, but relapse is common. Historically, treatment was limited to chemotherapy and radiotherapy [
93]. In recent years, the addition of immunotherapy has improved outcomes for selected patients (
Table 4).
4.3. Breast Cancer
Breast cancer is a heterogeneous group of malignant diseases that differ in histological, molecular, and clinical characteristics. Traditional classification is based on histology, with invasive ductal carcinoma being the most common type, while less frequent variants such as invasive lobular carcinoma and in situ lesions (ductal carcinoma in situ, lobular carcinoma in situ) are important for early diagnosis and monitoring [
94]. However, molecular classification—based on the expression of hormone receptors (estrogen and progesterone), HER2 receptor status, and the proliferation index (Ki-67)—is the key factor guiding systemic therapy and predicting disease outcomes [
95].
Based on these molecular features, breast cancer is commonly divided into four major subtypes: luminal A, luminal B, HER2-positive, and triple-negative breast cancer (TNBC). Luminal A tumors are characterized by strong hormone receptor expression with low proliferation, whereas luminal B tumors also express hormone receptors but have higher Ki-67 and/or HER2 positivity. The HER2-positive subtype is defined by HER2 overexpression or amplification in the absence of hormone receptors. Triple-negative cancers lack estrogen, progesterone, and HER2 receptors, and represent the most biologically aggressive group, prone to early metastasis and with limited therapeutic options [
96,
97].
Advances in molecular biology have enabled the identification of critical signaling pathways involved in breast cancer development and progression, leading to the introduction of targeted therapies. The most relevant therapeutic targets include the estrogen receptor pathway, the HER2 (ERBB2) pathway, cell cycle regulation via cyclins and cyclin-dependent kinases (CDKs), the PI3K/AKT/mTOR signaling axis, DNA repair mechanisms including BRCA mutations, as well as immunosuppressive pathways mediated by PD-1/PD-L1 interactions [
6,
56,
65,
66,
67,
68]. For patients with tumors that express the estrogen receptor, endocrine therapy remains the cornerstone of treatment. Selective estrogen receptor modulators (SERMs) such as tamoxifen and toremifene act as competitive antagonists of the receptor in breast tissue. Aromatase inhibitors, including anastrozole, letrozole, and exemestane, block peripheral estrogen synthesis, while selective estrogen receptor degraders (SERDs), such as fulvestrant and newer agents like elacestrant, directly induce receptor degradation [
67,
68]. In premenopausal patients, ovarian suppression can be achieved with LHRH agonists such as goserelin. HER2-positive breast cancers respond to monoclonal antibodies such as trastuzumab, pertuzumab, and newer derivatives like margetuximab [
98]. To further enhance efficacy, antibody–drug conjugates (ADCs) such as ado-trastuzumab emtansine and trastuzumab deruxtecan have been developed and are now widely used in clinical practice. Additionally, oral HER2 tyrosine kinase inhibitors—including lapatinib, neratinib, and tucatinib—provide intracellular blockade of HER2 signaling [
99]. CDK4/6 inhibitors (palbociclib, ribociclib, abemaciclib) have become the standard of care for HR+/HER2− metastatic breast cancer, in combination with endocrine therapy. These agents prevent cell cycle progression from the G1 to the S phase, thereby slowing tumor growth [
100]. Dysregulation of the PI3K/AKT/mTOR signaling pathway, particularly through PIK3CA mutations, is common in HR+ subtypes and is associated with resistance to endocrine therapy. PI3K inhibitors (alpelisib, inavolisib), the AKT inhibitor capivasertib, and the mTOR inhibitor everolimus represent therapeutic strategies in this setting [
101].
For triple-negative breast cancer, particularly those with BRCA1/2 mutations or other homologous recombination deficiencies, poly (ADP-ribose) polymerase (PARP) inhibitors such as olaparib and talazoparib are effective (
Figure 4) [
102]. Although tumors with homologous recombination deficiency initially respond to PARP inhibitors, resistance commonly develops through BRCA reversion mutations, epigenetic reactivation of DNA repair, loss of PARP1 function, and upregulation of drug efflux transporters. Understanding these resistance pathways, including mechanisms such as replication fork stabilization and engagement of alternative DNA repair processes, provides a foundation for designing combination therapies that can overcome resistance and improve the long-term efficacy of PARP inhibition.
In addition, immunotherapy with anti-PD-1 antibodies (pembrolizumab) has shown clinical benefit in patients whose tumors express PD-L1 [
103]. Newer strategies include antibody–drug conjugates targeting TROP2, such as sacituzumab govitecan and datopotamab deruxtecan, which are especially valuable for resistant forms of disease [
104].
All currently FDA-approved targeted therapies for breast cancer, along with their therapeutic targets, are summarized in
Table 5. These agents have significantly prolonged survival, improved quality of life, and advanced the concept of personalized oncology in clinical practice.
4.4. Colorectal Cancer
Colorectal cancer (CRC) is a biologically and clinically heterogeneous disease, with treatment increasingly guided by molecular profiling in addition to traditional staging. Molecular markers such as RAS (KRAS/NRAS) mutations, BRAF mutations, HER2 amplification, microsatellite instability (MSI) or mismatch repair (MMR) status, and tumor mutational burden (TMB) have become central to therapy selection and prognosis [
105]. The introduction of targeted therapies has transformed the management of metastatic CRC, expanding treatment beyond conventional chemotherapy. Key therapeutic strategies include targeting angiogenesis, inhibiting the EGFR pathway, blocking oncogenic drivers such as KRAS and BRAF, and harnessing immune checkpoint inhibition in biomarker-defined subgroups (
Table 6).
From a molecular perspective, CRC development and progression are driven by alterations in several major signaling pathways. The MAPK/ERK pathway, frequently activated by KRAS and BRAF mutations, promotes uncontrolled proliferation and survival, and is therapeutically targeted with EGFR inhibitors (cetuximab, panitumumab) in RAS wild-type tumors and with BRAF inhibitors (encorafenib in combination with cetuximab) in BRAF V600E–mutant disease [
4,
106,
107]. The PI3K/AKT/mTOR pathway, often altered through PIK3CA mutations or PTEN loss, regulates tumor growth and contributes to resistance against upstream inhibitors. WNT/β-catenin signaling, typically disrupted by APC mutations, is a hallmark of colorectal carcinogenesis, driving stemness and epithelial–mesenchymal transition, although direct pharmacologic targeting remains challenging [
48,
61]. In addition, angiogenesis signaling via VEGF/VEGFR plays a critical role in metastatic spread and is effectively targeted with agents such as bevacizumab, ramucirumab, ziv-aflibercept, and fruquintinib [
108]. Finally, immune pathways, particularly MMR deficiency and MSI-high status, identify subgroups highly responsive to immune checkpoint inhibitors such as pembrolizumab and nivolumab, with ipilimumab as a complementary CTLA-4 blockade [
109,
110].
4.5. Prostate Cancer
Prostate cancer is the most common malignancy among men in the United States and a leading cause of cancer-related death [
111]. Based on the latest research, the molecular mechanisms of prostate cancer (PCa) are complex and involve a variety of genetic, epigenetic, and signaling pathway alterations. Androgen Receptor (AR) Signaling is crucial in both the initiation and progression of PCa. Androgens bind to ARs, promoting transcription of genes that support cell survival and proliferation. However, in certain cases, despite androgen deprivation therapy (ADT), cancer could progress to Castration-Resistant Prostate Cancer (CRPC) due to: AR gene amplification, AR mutations and splice variants, intratumoral androgen synthesis or activation of AR by other signaling pathways. Genomic alterations play a crucial role in the development of PCa. There are common mutations in SPOP, FOXA1, TP53, PTEN, RB1, MYC, and other oncogene genes, while mutations linked to DNA repair defects are present at BRCA1/2 and ATM genes. On the other hand, Copy Number Alterations (CNAs) affect AR, MYC, and other oncogenes. Key signaling pathways involved in the development of PCa include PI3K/AKT/mTOR Pathway, Wnt/β-catenin Pathway, MAPK Pathways, and STAT3 and EZH2 Pathways, as well as alterations in cell cycle regulators, such as in CDK12 and RB1, that contribute to unchecked proliferation. Management has evolved significantly over the past decade, with the development of targeted therapies and next-generation hormonal agents that address key molecular drivers of disease progression (
Table 7). While androgen deprivation therapy (ADT) remains the cornerstone of treatment, new agents targeting the androgen receptor pathway, DNA repair defects, bone metastases, and other molecular vulnerabilities have expanded therapeutic options.
The development of targeted therapies for prostate cancer reflects the central role of the androgen receptor (AR) signaling pathway, along with the recognition of molecular vulnerabilities such as DNA repair deficiencies and prostate-specific antigens. One of the key advances has been the introduction of next-generation hormonal agents that directly or indirectly inhibit AR activity. Abiraterone acetate blocks androgen biosynthesis by inhibiting CYP17, while apalutamide, enzalutamide, and darolutamide act as potent AR antagonists, effectively suppressing tumor growth even in the castration-resistant state, particularly in metastatic castration-sensitive prostate cancer (mCSPC) [
112]. A comparative analysis of two androgen receptor inhibitors—enzalutamide (Xtandi) and darolutamide (Nubeqa)—showed that enzalutamide significantly delayed radiographic progression-free survival (rPFS) by nearly 50% compared to darolutamide in patients with metastatic hormone-sensitive prostate cancer (mHSPC) [
113]. This was based on a matching-adjusted indirect comparison (MAIC) of the ARCHES and ARANOTE trials.
Androgen deprivation therapy (ADT) remains a cornerstone of prostate cancer management, traditionally achieved through surgical castration or more commonly with LHRH agonists such as leuprolide, goserelin, and triptorelin [
114]. The GnRH antagonist degarelix provides a rapid and reversible method of testosterone suppression, while the oral agent relugolix offers a convenient alternative with similar efficacy. These approaches are essential in both localized high-risk disease and advanced metastatic settings, forming the foundation upon which newer therapies are layered [
115].
For patients with homologous recombination repair (HRR) gene mutations, PARP inhibitors have introduced a new level of precision. Olaparib, rucaparib, talazoparib, and the dual formulation of niraparib with abiraterone are approved options for men with BRCA1/2 or related DNA repair defects. These agents exploit synthetic lethality, selectively targeting cancer cells while sparing normal tissue [
116]. Finally, in patients who progress after androgen receptor-directed therapy and docetaxel, cabazitaxel, a next-generation taxane chemotherapy, provides meaningful benefit. Although not molecularly targeted in the classical sense, cabazitaxel is included among advanced prostate cancer treatments because of its specific activity against resistant disease [
117]. Theranostics combines diagnostics and therapy using radiopharmaceuticals that target prostate-specific membrane antigen (PSMA). The most notable agent is Pluvicto, which uses gallium-68 for imaging and lutetium-177 for treatment. Newer trials are exploring actinium-225, which emits more potent alpha particles for deeper tumor penetration [
118].
Kairos Pharma’s ENV105 is a resistance-reversing agent targeting CD105, a protein linked to treatment resistance. It is in Phase 2 trials for metastatic castration-resistant prostate cancer and shows promise in restoring the effectiveness of existing therapies like enzalutamide [
119].
4.6. Gastric Cancer
Gastric cancer remains a major global health challenge, often diagnosed at an advanced stage when curative surgery is not possible [
120]. Traditional chemotherapy regimens have long been the backbone of systemic treatment, but advances in molecular oncology and immunotherapy have expanded the therapeutic armamentarium (
Table 8). Today, targeted agents directed against HER2, angiogenesis pathways, Claudin 18.2, and immune checkpoints play a central role in the management of advanced gastric and gastroesophageal junction cancers [
121].
HER2 overexpression, present in a subset of gastric cancers, has provided one of the earliest and most impactful targets. Trastuzumab, a monoclonal antibody against HER2, is approved in combination with chemotherapy as the first targeted therapy to improve survival in HER2-positive gastric cancer. Building on this success, fam-trastuzumab deruxtecan, an antibody–drug conjugate, has demonstrated remarkable activity in patients who progress after trastuzumab-based therapy, offering a new line of effective treatment. Angiogenesis inhibition also has a defined role in gastric cancer. Ramucirumab, a monoclonal antibody targeting VEGFR2, is approved both as monotherapy and in combination with paclitaxel for patients with previously treated advanced disease [
98,
122]. Its approval was a major advance in the second-line setting, improving survival in a population with limited options.
In recent years, immunotherapy has reshaped gastric cancer treatment. PD-1 inhibitors, including nivolumab, pembrolizumab, and, more recently tislelizumab, are approved in biomarker-selected populations. Nivolumab has shown benefit in combination with chemotherapy in HER2-negative advanced gastric cancer, while pembrolizumab is used in PD-L1-positive tumors and microsatellite instability-high (MSI-H) cancers [
123]. Tislelizumab, a PD-1 inhibitor recently approved, further expands the options available for immune checkpoint blockade in this disease. Another important breakthrough is the development of zolbetuximab, the first-in-class antibody targeting Claudin 18.2, a tight junction protein expressed in a subset of gastric and gastroesophageal junction cancers. Approved in combination with chemotherapy, zolbetuximab offers a novel biomarker-driven therapy for patients with CLDN18.2-positive tumors, marking a new step toward personalized treatment in gastric cancer [
124]. Durvalumab (Imfinzi), an anti-PD-L1 monoclonal antibody, has shown significant promise in treating resectable gastric and gastroesophageal junction (GEJ) cancers, based on the results of the MATTERHORN Phase III trial presented at ASCO 2025. MATTERHORN Phase III trial included 948 patients with stage II–IVA resectable gastric or GEJ adenocarcinoma who were randomized to receive either: Durvalumab + FLOT chemotherapy (perioperative: 2 neoadjuvant + 2 adjuvant cycles, followed by 10 cycles of durvalumab monotherapy), Placebo + FLOT, followed by placebo maintenance. Event-Free Survival (EFS) 24-month EFS: 67.4% (durvalumab) vs. 58.5% (placebo), while Hazard Ratio: 0.71 (95% CI: 0.58–0.86),
p < 0.001 [
125]. FDA Priority Review & Breakthrough Therapy Designation was granted for Durvalumab in July 2025, and approval decision is expected in Q4 2025. Durvalumab combined with FLOT chemotherapy is poised to redefine the standard of care for resectable gastric and GEJ cancers. It is the first immunotherapy regimen to show statistically significant improvements in EFS and Pathologic Complete Response (pCR) in this setting [
126].
4.7. Liver and Bile Duct Cancers
Primary liver cancers, most commonly hepatocellular carcinoma (HCC), and bile duct cancers (cholangiocarcinomas) are aggressive malignancies often diagnosed at advanced stages [
127]. For many years, systemic treatment options were limited, with sorafenib as the only approved therapy. However, the past decade has seen remarkable progress with the introduction of targeted therapies and immunotherapies that have redefined the standard of care (
Table 9).
In hepatocellular carcinoma, angiogenesis plays a critical role in tumor biology, and VEGF pathway inhibitors have been central to treatment. Sorafenib, the first agent to demonstrate survival benefit, remains an option, while lenvatinib, regorafenib, and cabozantinib offer alternatives or second-line therapy. Bevacizumab, an anti-VEGF antibody, in combination with the PD-L1 inhibitor atezolizumab, has become a frontline standard, providing superior survival compared with sorafenib. Ramucirumab, targeting VEGFR2, is another important therapy in biomarker-selected patients with elevated alpha-fetoprotein (AFP) [
128].
Checkpoint inhibitors have transformed liver cancer management, particularly in immunotherapy-based combinations. Nivolumab and pembrolizumab are PD-1 inhibitors with established roles in HCC, while durvalumab, another PD-L1 inhibitor, is approved in combination with the CTLA-4 inhibitor tremelimumab. This dual checkpoint strategy enhances antitumor immunity and has provided durable responses for a subset of patients. Ipilimumab and tremelimumab, in combination with PD-1/PD-L1 agents, further expand the immunotherapy landscape in advanced liver cancer [
129].
For bile duct cancers, precision oncology has played a transformative role. FGFR2 fusions are actionable targets in intrahepatic cholangiocarcinoma, with pemigatinib and futibatinib now approved in this setting. IDH1 mutations, present in a subset of cholangiocarcinomas, can be treated with ivosidenib, providing an additional biomarker-driven approach. More recently, zanidatamab, a HER2-targeted bispecific antibody, has been approved for HER2-amplified biliary tract cancers, addressing another clinically relevant molecular subset [
130].
4.8. Thyroid Cancer
Thyroid cancer encompasses a heterogeneous group of malignancies, including differentiated thyroid cancers (papillary, follicular, and Hurthle cell), medullary thyroid cancer, and the aggressive anaplastic thyroid carcinoma. While surgery and radioactive iodine remain the cornerstones of therapy for most differentiated thyroid cancers, a subset of patients develop advanced or refractory disease that no longer responds to traditional approaches [
131]. In these cases, targeted therapies have become essential, providing meaningful clinical benefit through inhibition of specific molecular pathways (
Table 10).
The vascular endothelial growth factor (VEGF) signaling pathway is a central driver of thyroid tumor growth and angiogenesis. Multikinase inhibitors such as sorafenib and lenvatinib have become standard options for radioactive iodine–refractory differentiated thyroid cancer, significantly prolonging progression-free survival. Cabozantinib, another multikinase inhibitor with activity against VEGFR, MET, and RET, is approved for medullary thyroid cancer and as a second-line option in differentiated thyroid cancer. Vandetanib similarly targets VEGFR, EGFR, and RET and is approved for advanced medullary thyroid cancer, providing durable disease control in this rare subtype [
132,
133].
The discovery of actionable genetic alterations has further refined thyroid cancer therapy. RET fusions and mutations, common in medullary and some papillary thyroid cancers, can be effectively treated with selective RET inhibitors such as selpercatinib and pralsetinib, which have demonstrated high response rates and favorable tolerability compared to older multikinase inhibitors. For patients with anaplastic thyroid carcinoma harboring the BRAF V600E mutation, the combination of dabrafenib (a BRAF inhibitor) and trametinib (a MEK inhibitor) provides an important targeted option, dramatically improving outcomes in a disease that previously had a dismal prognosis [
134,
135].
4.9. Bladder Cancer
Bladder cancer, most often presenting as urothelial carcinoma, is a complex malignancy historically treated with surgery, platinum-based chemotherapy, and intravesical therapy for early-stage disease [
126]. In recent years, advances in molecular biology and immuno-oncology have led to the approval of several targeted therapies and immunotherapies (
Table 11), particularly for patients with advanced or metastatic disease who progress after standard chemotherapy. These treatments have substantially expanded the therapeutic landscape, providing durable responses and improving survival in selected patient populations [
136].
A breakthrough has been the introduction of immune checkpoint inhibitors, which harness the body’s own immune system to fight cancer. Agents such as atezolizumab, avelumab, nivolumab, and pembrolizumab target the PD-1/PD-L1 pathway, restoring T-cell activity against tumor cells. These therapies are approved for advanced urothelial carcinoma in patients who have failed prior chemotherapy, and in some cases as first-line treatment for cisplatin-ineligible patients with PD-L1-positive tumors [
137]. Avelumab also has a unique role as maintenance therapy after first-line platinum-based chemotherapy, prolonging overall survival compared with best supportive care.
In addition to checkpoint blockade, antibody–drug conjugates have become an important treatment option. Enfortumab vedotin, directed against Nectin-4 and linked to a cytotoxic agent, has demonstrated meaningful activity in patients previously treated with chemotherapy and immunotherapy. Its approval has been a major step forward for those with limited remaining treatment options. Targeted small-molecule inhibitors also contribute to precision therapy in bladder cancer. Erdafitinib, an oral fibroblast growth factor receptor (FGFR) inhibitor, is approved for tumors harboring susceptible FGFR2 or FGFR3 alterations. By blocking aberrant FGFR signaling, erdafitinib provides benefit in a biomarker-defined population that historically had poor responses to standard therapy [
138]. More recently, immunotherapy innovation has expanded further with nogapendekin alfa inbakicept, an IL-15 superagonist designed to enhance natural killer and T-cell activity [
139]. Its approval for BCG-unresponsive non-muscle invasive bladder cancer represents an important alternative for patients who might otherwise require radical surgery.
4.10. Renal Cell Carcinoma
Renal cell carcinoma (RCC), has historically been resistant to conventional chemotherapy and radiation [
140]. Over the past two decades, a deeper understanding of tumor biology—particularly angiogenesis, hypoxia-inducible pathways, and immune evasion—has led to the development of multiple targeted therapies and immunotherapies that have transformed patient outcomes (
Table 12). These agents, used either as monotherapy or in combination, form the backbone of treatment for advanced and metastatic RCC [
140].
One of the earliest therapeutic breakthroughs was the inhibition of angiogenesis, a hallmark of RCC. Agents such as bevacizumab, an anti-VEGF antibody, and multiple VEGF receptor tyrosine kinase inhibitors (TKIs), including sunitinib, sorafenib, pazopanib, axitinib, cabozantinib, lenvatinib, and tivozanib, have demonstrated robust clinical activity. These drugs block the VEGF signaling cascade, reducing blood supply to tumors and slowing progression. Their development marked a turning point in kidney cancer therapy, shifting the standard of care away from nonspecific immunotherapies such as interferon. Beyond VEGF inhibition, additional pathways have been successfully targeted [
140,
141]. The hypoxia-inducible factor 2α (HIF-2α) inhibitor belzutifan provides a novel option for patients with von Hippel–Lindau disease-associated RCC and has expanded into broader RCC indications [
142]. Immunotherapy has also revolutionized kidney cancer management. The checkpoint inhibitors nivolumab and pembrolizumab, targeting PD-1, have shown durable responses, while ipilimumab, a CTLA-4 inhibitor, is used in combination with PD-1 blockade to enhance T-cell activity. Avelumab, a PD-L1 inhibitor, also plays a role in combination regimens with TKIs such as axitinib [
140,
143]. These immune checkpoint inhibitors have redefined the therapeutic landscape, providing long-term disease control for a subset of patients previously considered incurable.
4.11. Leukemias
Leukemia is a heterogeneous group of hematologic malignancies arising from the bone marrow and blood. Treatment strategies vary by disease subtype—acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myeloid leukemia (CML), and chronic lymphocytic leukemia (CLL)—and are increasingly guided by molecular biomarkers and immunophenotyping [
144]. The introduction of targeted therapies, including tyrosine kinase inhibitors (TKIs), monoclonal antibodies, antibody–drug conjugates, small-molecule inhibitors, and cellular immunotherapies, has transformed outcomes across leukemias (
Table 13). The introduction of targeted therapies has revolutionized the treatment of leukemias, shifting the focus from conventional cytotoxic chemotherapy to precision medicine tailored to molecular drivers and immune characteristics. In chronic myeloid leukemia (CML), the discovery of the BCR-ABL fusion gene established tyrosine kinase inhibitors as the standard of care. Imatinib was the first to transform outcomes, and it has since been followed by more potent agents such as dasatinib, nilotinib, bosutinib, ponatinib, and the allosteric inhibitor asciminib, which address resistance mutations, including T315I [
145]. In acute myeloid leukemia (AML), a different spectrum of targeted therapies has emerged, particularly for patients with FLT3 mutations [
146]. Agents such as midostaurin, gilteritinib, and quizartinib inhibit aberrant FLT3 signaling, while enasidenib, ivosidenib, and olutasidenib target IDH2 and IDH1 mutations, restoring normal differentiation pathways. The menin inhibitor revumenib represents an important advance for KMT2A-rearranged and NPM1-mutated leukemias, further broadening the therapeutic landscape [
147,
148].
Differentiation therapy remains a cornerstone for specific AML subsets, most notably acute promyelocytic leukemia, where tretinoin (all-trans retinoic acid) induces maturation of malignant promyelocytes [
149]. Additional agents such as glasdegib, which targets the Hedgehog signaling pathway, further reflect the trend toward pathway-directed strategies in AML [
150]. In parallel, therapies directed at B-cell surface antigens have dramatically improved the outlook for lymphoid leukemias. Anti-CD20 antibodies, including rituximab, obinutuzumab, and ofatumumab, are central in chronic lymphocytic leukemia (CLL), while alemtuzumab, which targets CD52, remains an option for refractory disease [
151].
Cellular immunotherapy has opened an entirely new chapter in the treatment of leukemias. Chimeric antigen receptor (CAR) T-cell therapies such as tisagenlecleucel, brexucabtagene autoleucel, lisocabtagene maraleucel, and obecabtagene autoleucel harness the patient’s own immune system to recognize and eradicate CD19-expressing blasts in relapsed or refractory ALL and certain lymphoid leukemias. Complementing these, tagraxofusp, a CD123-directed fusion protein, addresses rare leukemic presentations such as blastic plasmacytoid dendritic cell neoplasm [
152,
153].
4.12. Pancreatic Cancer
Pancreatic cancer, particularly pancreatic ductal adenocarcinoma (PDAC), remains one of the most lethal malignancies, with limited curative options and a poor overall prognosis [
154]. While cytotoxic chemotherapy continues to serve as the cornerstone of treatment, advances in molecular profiling have revealed actionable alterations in a subset of patients, enabling the use of targeted therapies (
Table 14). These developments represent an important step toward individualized care in a disease long resistant to therapeutic advances.
One of the earliest targeted agents approved for pancreatic cancer is erlotinib, an EGFR tyrosine kinase inhibitor, used in combination with gemcitabine. Although the survival benefit is modest, it marked the beginning of targeted approaches in this cancer type [
155]. For patients with germline BRCA1/2 mutations, the PARP inhibitor olaparib has changed clinical practice. Approved as maintenance therapy for metastatic disease following platinum-based chemotherapy, olaparib leverages synthetic lethality to exploit DNA repair deficiencies, extending survival in biomarker-selected patients [
156]. Everolimus, an mTOR inhibitor, and sunitinib, a multikinase inhibitor, are primarily approved for pancreatic neuroendocrine tumors (pNETs) rather than pancreatic adenocarcinoma. Both agents significantly improve progression-free survival and remain key options in the management of advanced pNETs, highlighting the importance of distinguishing tumor histologies within the pancreas [
157]. More recent advances include belzutifan, a HIF-2α inhibitor approved for von Hippel–Lindau (VHL)-associated pancreatic neuroendocrine tumors, further expanding the precision oncology toolbox in hereditary cancer syndromes [
158]. Additionally, zenocutuzumab, a bispecific antibody targeting HER2 and HER3, is approved for tumors with NRG1 fusions, a rare but actionable alteration occasionally found in pancreatic cancer [
159].
Although the proportion of patients eligible for these therapies remains relatively small, the approval of EGFR, PARP, VEGF/mTOR pathway inhibitors, and NRG1-targeted therapies demonstrates a growing ability to personalize treatment. Ongoing research into KRAS inhibitors, immunotherapy combinations, and other novel approaches promises to further broaden the scope of targeted therapy in pancreatic cancer.
4.13. Brain Tumors
Brain cancer represents a heterogeneous group of malignant diseases that differ in histological, molecular, and clinical features. While traditional classification relied primarily on histology—such as glioblastoma (GMB), oligodendroglioma, and astrocytoma—modern diagnostic standards now place molecular characteristics at the center of glioma classification [
34]. In fact, the current WHO classification of central nervous system tumors mandates integration of molecular markers, including IDH1/2 mutations, 1p/19q codeletion status, ATRX and TP53 alterations, and MGMT promoter methylation, as essential criteria for accurate diagnosis. In some instances, such as differentiating oligodendroglioma grade 3 from glioblastoma, molecular testing (e.g., fluorescence in situ hybridization, FISH, for 1p/19q codeletion) is indispensable, as histology alone is insufficient to distinguish between entities with profoundly different prognosis and treatment strategies [
34,
160]. Advances in molecular biology have further enabled the identification of critical signaling pathways involved in tumorigenesis and progression of brain cancers, leading to the development of targeted therapies. Among the most important therapeutic targets are hypoxia-inducible factor (HIF) pathways, angiogenesis mediated by vascular endothelial growth factor (VEGF), MAPK signaling, the mTOR pathway, and mutant metabolic enzymes such as IDH1/2 [
34,
161,
162,
163].
Several targeted agents have been approved for the treatment of brain tumors, particularly in recurrent or molecularly defined settings (
Table 15). These agents have significantly expanded the therapeutic landscape by allowing for more personalized treatment approaches. While traditional chemotherapy and radiotherapy remain the backbone of therapy, targeted treatments provide meaningful benefits in tumor control, progression-free survival, and quality of life, particularly in biomarker-driven subgroups.
EGFR inhibitors (e.g., erlotinib, afatinib) and EGFRvIII-targeted vaccines have shown limited success in glioblastoma patients, due to poor BBB penetration and intratumoral heterogeneity. At the same time, Bevacizumab, an anti-VEGF monoclonal antibody, is FDA-approved for recurrent GBM. It improves progression-free survival but not overall survival. On the other hand, Vorasidenib, an oral Isocitrate Dehydrogenase (IDH1/2) inhibitor, recently approved for IDH-mutant gliomas, significantly prolongs progression-free survival [
164].
4.14. Skin Cancers
Skin cancers represent a diverse group of malignancies that include melanoma, cutaneous squamous cell carcinoma, basal cell carcinoma, and rare entities such as uveal melanoma and cutaneous T-cell lymphoma [
165]. While surgery and radiotherapy remain central for localized disease, the management of advanced and metastatic skin cancers has been revolutionized by targeted therapies and immunotherapies (
Table 16). These agents exploit specific oncogenic drivers or modulate immune checkpoint pathways, leading to durable responses in patients with historically limited options.
In melanoma, the discovery of activating mutations in the BRAF gene (V600E/K) has transformed treatment. BRAF inhibitors such as vemurafenib, dabrafenib, and encorafenib, particularly when combined with MEK inhibitors like trametinib, cobimetinib, or binimetinib, provide robust and durable responses while delaying resistance [
166]. For patients without BRAF mutations, immune checkpoint inhibitors, including anti-PD-1 antibodies (nivolumab, pembrolizumab, cemiplimab, cosibelimab, retifanlimab) and CTLA-4 inhibition (ipilimumab), have redefined outcomes, either as monotherapy or in combinations such as nivolumab–relatlimab (targeting PD-1 and LAG-3) [
143]. Cutaneous squamous cell carcinoma (cSCC) has shown remarkable sensitivity to PD-1 inhibitors, with cemiplimab and pembrolizumab approved as standard therapies [
167]. These agents highlight the profound role of immunotherapy in non-melanoma skin cancers. Basal cell carcinoma (BCC), driven in many cases by aberrant Hedgehog pathway signaling, is effectively treated with smoothened (SMO) inhibitors such as vismodegib and sonidegib [
168]. These drugs directly target the molecular drivers of tumor growth, representing a precision approach in advanced BCC resistant to local therapy.
For rare skin malignancies, specialized agents have emerged. Alitretinoin, a retinoid, is used for cutaneous T-cell lymphoma [
169]. Tebentafusp, a bispecific fusion protein targeting gp100 and CD3, is approved for uveal melanoma, marking the first therapy to demonstrate an overall survival benefit in this rare disease [
170]. Additionally, avelumab and atezolizumab have roles in Merkel cell carcinoma, a highly aggressive but immunotherapy-sensitive skin cancer [
171].
4.15. Ovarian Epithelial, Fallopian Tube, and Primary Peritoneal Cancers
Ovarian epithelial cancers, along with closely related fallopian tube and primary peritoneal cancers, represent the majority of gynecologic malignancies in this category [
172]. These cancers are often diagnosed at an advanced stage, and while surgery and platinum-based chemotherapy remain central components of treatment, the development of targeted therapies has significantly changed the therapeutic landscape (
Table 17). Precision medicine approaches now allow clinicians to tailor therapy based on molecular alterations and tumor biology, improving both survival and quality of life.
One of the most impactful advances has been the introduction of PARP inhibitors for tumors with homologous recombination deficiency (HRD), including BRCA1/2 mutations. Olaparib, niraparib, and rucaparib are approved for maintenance therapy following chemotherapy response, as well as for recurrent disease in biomarker-selected populations. These agents exploit synthetic lethality, blocking DNA repair pathways and leading to selective tumor cell death in HRD-positive cancers. Their integration into frontline and recurrent settings has transformed the long-term management of ovarian cancer [
173]. Another important therapeutic class is angiogenesis inhibition. Bevacizumab, a VEGF-targeting monoclonal antibody, is approved both as maintenance therapy in the frontline setting and as part of treatment for recurrent disease [
141]. By disrupting tumor vascular supply, bevacizumab slows disease progression and provides additional benefit when combined with chemotherapy.
A more recent innovation is the use of antibody–drug conjugates (ADCs). Mirvetuximab soravtansine, an ADC targeting folate receptor alpha (FRα), was recently approved for platinum-resistant ovarian cancer with high FRα expression [
174]. This agent delivers a cytotoxic payload directly into tumor cells, combining targeted precision with potent antitumor activity, and provides an important new option for patients with limited therapeutic alternatives.