Introduction to Anlotinib Dihydrochloride
Anlotinib Dihydrochloride is a novel oral small molecule tyrosine kinase inhibitor (TKI) that has been developed to target multiple signaling pathways involved in tumor growth and angiogenesis. As a molecular-targeted therapeutic agent, it exerts its antitumor activity primarily by interfering with aberrant receptor signaling pathways that are critical for tumor proliferation and vascular development. Developed originally in China and approved in clinical settings for advanced non‐small cell lung cancer (NSCLC) among other malignancies, anlotinib is recognized for its potent inhibition of various receptor tyrosine kinases and its broad-spectrum antitumor efficacy.
Chemical and Pharmacological Profile
Anlotinib is categorized as a small molecule drug with a unique chemical structure that allows it to bind with high affinity to several receptor sites on target proteins. Its structure is designed to fit within the adenosine triphosphate (ATP) binding pocket of multiple receptor tyrosine kinases, thereby competitively inhibiting ATP binding and subsequent phosphorylation of downstream signaling molecules. This molecular design lends anlotinib dual properties: it not only disrupts intracellular signal transduction pathways essential for cell proliferation but also significantly impairs angiogenic signaling cascades. In pharmacological studies, anlotinib has demonstrated a high degree of specificity for its primary targets, including vascular endothelial growth factor receptors (VEGFR1, VEGFR2, VEGFR3), fibroblast growth factor receptors (FGFRs), platelet-derived growth factor receptors (PDGFR α/β), and c-Kit. The drug’s favorable pharmacokinetic profile results in adequate oral bioavailability, good tissue penetration (including in tumor tissues), and rapid absorption, which enable sustained inhibition of kinase activity at clinically relevant doses.
Therapeutic Indications
From a therapeutic standpoint, anlotinib has been extensively studied and has demonstrated significant efficacy in treating a range of malignancies. Its principal indication includes advanced non‐small cell lung cancer, where it has already been approved as a third-line treatment option. Beyond NSCLC, clinical studies have investigated anlotinib’s benefit in cancers such as soft tissue sarcoma, renal cell carcinoma, medullary thyroid carcinoma, colorectal cancer, and even osteosarcoma. The broad targeting profile of anlotinib—addressing both tumor cell proliferation and angiogenesis—makes it a promising candidate not only as a monotherapy but also in combination with other treatment modalities, including chemotherapy and immunotherapy. Its ability to target tumor vascular networks gives it a particularly valuable role in combating tumors that exhibit a high level of angiogenic activity, which is a hallmark of aggressive and refractory cancers.
Molecular Mechanism of Action
The molecular mechanism of action of anlotinib Dihydrochloride is multifaceted. At its core, the drug inhibits the kinase activities of a broad spectrum of receptor tyrosine kinases (RTKs) that are intimately involved in both tumor cell survival and angiogenesis. This blocking of signaling pathways leads to the suppression of tumor proliferation, angiogenic vessel formation, and ultimately tumor growth.
Target Receptors and Pathways
Anlotinib exerts its biological effects through high-affinity interactions with several key receptor tyrosine kinases (RTKs). Its primary targets include:
• VEGFR1, VEGFR2, and VEGFR3 – These receptors are central to the vascular endothelial growth factor (VEGF)-dependent angiogenic processes. By blocking VEGFR signaling, anlotinib interferes with the formation, maintenance, and permeability of blood vessels that sustain tumor growth and metastasis.
• FGFRs – The fibroblast growth factor receptors are implicated in cell proliferation, differentiation, and angiogenesis. Inhibition of FGFR signaling by anlotinib disrupts pathways crucial for cell survival and vascular development in the tumor microenvironment, thereby impeding the tumor’s ability to secure a supportive blood supply.
• PDGFR α/β – Platelet-derived growth factor receptors play an essential role in the maturation and stabilization of blood vessels. Targeting PDGFRs disrupts pericyte recruitment and vessel stabilization, contributing further to the antiangiogenic effects of anlotinib.
• c-Kit – This receptor is involved in cellular signaling cascades that support cell differentiation and proliferation. Inhibition of c-Kit may additionally contribute to the suppression of tumor growth in malignancies where c-Kit is overexpressed.
The inhibition of these kinases results in the disruption of multiple downstream signaling cascades. For instance, blocking VEGFR engagement prevents the activation of key pathways such as the PI3K/AKT/mTOR and the RAS/RAF/MEK/ERK cascades, which are normally responsible for promoting cell survival, migration, and proliferation. This multi-targeted approach not only directly affects tumor cell viability but also hampers the tumor’s ability to sustain and remodel its microenvironment.
Inhibition of Angiogenesis
Angiogenesis, defined as the formation of new blood vessels from preexisting vasculature, is a fundamental process that tumors exploit to provide nutrients and oxygen as they grow. Anlotinib’s antiangiogenic properties are primarily due to its potent inhibition of the VEGF/VEGFR signaling axis. By binding to VEGFR1, VEGFR2, and VEGFR3, anlotinib prevents the receptor activation that is necessary for initiating the angiogenic cascade.
In addition, inhibition of FGFR and PDGFR signaling further contributes to its ability to arrest new vessel formation. Tumor endothelial cells, which normally rely on these signals for proliferation and migration, become functionally impaired in the presence of anlotinib. This leads to a reduction in microvessel density and normalization of the abnormal tumor vasculature. The overall impact is a marked decrease in tumor blood flow, which starves the tumor of essential nutrients and oxygen, thus hindering tumor growth and metastasis.
Recent studies have shown that anlotinib can induce long-term vessel normalization in tumors. This normalization not only contributes to direct antitumor effects by limiting angiogenesis but also potentially improves the delivery of co-administered chemotherapy agents. Furthermore, the inhibition of angiogenesis by anlotinib is associated with a significant decrease in the expression of angiogenic markers, such as CD31, which has been used as a surrogate measure of microvessel density in tumor tissues.
Cellular and Molecular Effects
Beyond its broad-spectrum inhibition of receptor tyrosine kinases, anlotinib exerts a series of cellular and molecular effects that culminate in tumor growth suppression and enhanced antitumor immunity. These effects are observed in both the tumor cells themselves and in the surrounding vascular endothelial cells, which are critical for the maintenance of the tumor microenvironment.
Impact on Tumor Cells
Anlotinib directly affects tumor cells through several mechanisms:
• Inhibition of Proliferation and Cell Cycle Arrest:
Anlotinib has been shown to induce cell cycle arrest – particularly at the G2/M phase – in tumor cells such as those derived from thyroid cancer, oral squamous cell carcinoma, and colorectal cancer. By halting the progression of the cell cycle, the drug limits the ability of cancer cells to proliferate. This effect is often accompanied by changes in cell cycle regulatory proteins, including upregulation of cell cycle inhibitors like p21 and downregulation of proliferative markers such as Ki67.
• Induction of Apoptosis and Autophagy:
Studies have consistently demonstrated that treatment with anlotinib results in the induction of apoptosis in multiple cancer cell types. This pro-apoptotic effect involves both intrinsic and extrinsic pathways. For instance, apoptotic induction is associated with a decrease in the expression of anti-apoptotic proteins (e.g., Bcl-2) and an increase in pro-apoptotic mediators (e.g., Bax, cleaved caspase-3). Moreover, evidence indicates that anlotinib may promote cell autophagy in certain malignancies, contributing to its overall cytotoxic effects. In breast cancer cell lines, autophagy induction has been linked to the inhibition of the Akt/GSK-3α pathway, ultimately sensitizing cells to apoptosis.
• Suppression of Invasion and Metastasis:
By interfering with key signaling pathways such as the MEK/ERK cascade, anlotinib reduces the migratory and invasive capabilities of tumor cells. This has been demonstrated in both in vitro assays (e.g., wound-healing, transwell migration assays) and in vivo models, where treatment with anlotinib leads to reduced metastatic spread. Notably, anlotinib’s activity against KRAS-mutated lung cancer cells involves the suppression of the MEK/ERK pathway, indicating its potential efficacy in recalcitrant tumor types where conventional therapies have failed.
• Direct Targeting of Oncogenic Drivers:
Recent findings have suggested that anlotinib can directly target critical oncogenic molecules such as c-Myc. The enhanced ubiquitin-proteasomal degradation of c-Myc following anlotinib treatment contributes to tumor cell apoptosis and, consequently, a reduction in cell proliferation. This additional mechanism underscores the drug’s capacity to impact tumor cells at multiple regulatory levels.
Effects on Vascular Endothelial Cells
The hallmark antiangiogenic action of anlotinib is reflected in its direct impact on vascular endothelial cells:
• Inhibition of Endothelial Cell Proliferation and Tube Formation:
Anlotinib interferes with the ability of endothelial cells to undergo proliferation, migration, and tube formation—the fundamental processes required for new blood vessel formation. In vitro studies using human umbilical vein endothelial cells (HUVECs) have shown that anlotinib treatment results in a dose-dependent inhibition of these cellular processes. The drug’s effectiveness in reducing tube formation is indicative of its potential to disrupt the vascular network that sustains tumor growth.
• Reduction of Microvessel Density in Tumors:
In animal models, anlotinib induces a significant decrease in microvessel density, as evidenced by reduced CD31 expression in tumor tissues. This reduction is a direct consequence of the blockade of VEGFR, FGFR, and PDGFR signaling, impairing the recruitment and proliferation of endothelial cells within the tumor microenvironment.
• Vascular Normalization and Improved Drug Delivery:
Interestingly, beyond simply pruning tumor vessels, anlotinib has been reported to induce a phenomenon known as vascular normalization. This process transforms the abnormal, highly permeable and disorganized tumor vasculature into a more structured and functional network, thereby enhancing perfusion. Such normalization is thought to improve the delivery of chemotherapeutic and immunotherapeutic agents to the tumor site while also reducing hypoxia—a condition that often contributes to treatment resistance.
• Suppression of Endothelial Signaling Cascades:
At the molecular level, anlotinib diminishes the phosphorylation of critical endothelial signaling proteins, notably within the PI3K/AKT pathway. This attenuation further disrupts cell survival signals in endothelial cells, reinforcing the drug’s antiangiogenic potential and leading to the collapse of the tumor’s vascular support system.
Clinical Implications
The molecular and cellular actions of anlotinib translate into significant clinical benefits. Its ability to exert direct cytotoxic effects on tumor cells in conjunction with its powerful antiangiogenic properties is central to its clinical efficacy in a variety of cancer types.
Efficacy in Different Cancer Types
Clinical trials and preclinical studies have consistently demonstrated that anlotinib is effective against multiple tumor types:
• Non–Small Cell Lung Cancer (NSCLC):
One of the major breakthrough indications for anlotinib is in advanced NSCLC. Phase III clinical trials have demonstrated that patients treated with anlotinib experience a marked improvement in progression-free and overall survival, particularly in those who have progressed after two or more lines of prior treatment.
• Soft Tissue Sarcoma and Hepatocellular Carcinoma:
In soft tissue sarcoma models, anlotinib has shown significant antitumor activity, with clinical trials reporting prolonged disease control. Its antitumor effects have also been observed in hepatocellular carcinoma, where the inhibition of VEGFR and FGFR pathways plays a pivotal role.
• Colorectal Cancer and Ovarian Cancer:
Early research and clinical protocols have focused on the application of anlotinib in colorectal cancer, where it appears to reduce cell proliferation, induce apoptosis, and diminish tumor microvessel density, ultimately suppressing tumor growth. Similarly, in ovarian cancer, anlotinib has been implicated in inducing G2/M phase arrest and apoptosis, thereby offering a promising therapeutic option.
• Other Malignancies (e.g., Thyroid Cancer, Oral Squamous Cell Carcinoma, Osteosarcoma):
Anlotinib’s broad target profile means that it holds promise for a variety of other tumor types. In thyroid cancer, for example, it can reduce tumor burden even in cases that are initially considered unresectable. In oral squamous cell carcinoma, anlotinib’s ability to induce apoptosis and mitotic catastrophe offers significant therapeutic potential. In osteosarcoma models, the dual blockade of VEGFR2 and MET by anlotinib has demonstrated both anti-proliferative and antimetastatic effects.
Combination Therapies
Given its multi-targeted mechanism, anlotinib is not only effective as monotherapy but also enhances the efficacy of combination treatment strategies. There are several dimensions to its use in combination therapies:
• Chemotherapy Combinations:
Anlotinib has been combined with cytotoxic chemotherapy agents to increase antitumor efficacy. When used in combination regimens, particularly in gastrointestinal (GI) tumors and lung cancers, anlotinib has been shown to improve objective response rates and prolong progression-free survival compared to monotherapy. The antiangiogenic properties of anlotinib complement the cell kill achieved by chemotherapy, while also potentially normalizing tumor vasculature to enhance drug delivery.
• Immunotherapy Combinations:
The evolving landscape of cancer treatment has seen the incorporation of immune checkpoint inhibitors. Studies have demonstrated that low-dose anlotinib treatment can induce long-term tumor vascular normalization, thereby creating a microenvironment that is more conducive to immune cell infiltration. When combined with PD-1 inhibitors (e.g., camrelizumab) in breast tumor models, the combination has led to improved antitumor responses with increased percentages of intratumoral CD4+ T cells, CD8+ T cells, and natural killer (NK) cells. Combination therapy in this context exploits both the antiangiogenic effect of anlotinib and the immune-mediated tumor cell killing by checkpoint inhibition, offering a dual-pronged strategy against cancer.
• Targeted Therapy Combinations:
Considering the multi-targeted action of anlotinib, its combination with other receptor-specific targeted agents can yield synergistic effects. For instance, combining anlotinib with drugs that target compensatory or parallel pathways may overcome resistance mechanisms and potentiate antitumor activity. This rational design is based on an improved understanding of the underlying signaling networks in tumors, and several clinical trials are currently investigating such combinations.
Challenges and Future Directions
While the clinical efficacy of anlotinib is well documented, several challenges and areas for future research remain in order to fully optimize its use in cancer therapy.
Resistance Mechanisms
Despite its broad target range and clinical benefits, resistance to anlotinib, as with many targeted therapies, can develop over time. Some of the key challenges associated with resistance include:
• Tumor Adaptation and Alternate Angiogenic Pathways:
Tumors have the inherent ability to adapt to antiangiogenic therapies by upregulating alternative proangiogenic factors. In the context of anlotinib therapy, redundancy in signaling pathways (e.g., secretion of alternate growth factors that can signal through non-targeted receptors) may diminish its efficacy over time.
• Genetic and Epigenetic Alterations:
Long-term treatment can lead to genetic mutations or epigenetic modifications in tumor cells that confer resistance. For example, alterations in the expression or function of the target receptors, or changes in downstream signaling components, can reduce the sensitivity of tumor and endothelial cells to anlotinib.
• Tumor Microenvironment Changes:
The dynamic nature of the tumor microenvironment may also contribute to resistance. Changes in stromal cell interactions, extracellular matrix remodeling, and shifts in cytokine profiles can all impact the drug’s effectiveness. Understanding these adaptive mechanisms is critical for developing strategies to prevent or overcome resistance.
Ongoing Research and Development
To address these challenges, ongoing research is focused on several fronts:
• Biomarker Identification:
Effective biomarkers that predict response to anlotinib are urgently needed. Such biomarkers would help tailor treatment regimens to individual patients, thereby optimizing therapeutic outcomes while minimizing unnecessary toxicity. Current research is exploring various genomic, proteomic, and metabolomic markers to guide patient selection and dosing regimens.
• Improved Combination Strategies:
New combination therapies are under investigation to overcome resistance by simultaneously targeting multiple pathways or by adding agents that modulate the tumor microenvironment. Clinical trials combining anlotinib with immunotherapies, chemotherapeutic agents, and other targeted drugs are ongoing, with early-phase results suggesting improved efficacy and manageable toxicity profiles.
• Mechanistic Studies Using Systems Biology:
Advanced systems biology approaches and network modeling are being used to understand the intricate signaling networks influenced by anlotinib. These investigations provide insights into secondary targets and off-target effects, enabling the rational design of next-generation combination therapies. Such studies are crucial for understanding the drug’s impact on both cancer cells and the surrounding stroma, thereby refining therapeutic strategies.
• Dose Optimization and Scheduling:
Clinical studies are also exploring different dosing regimens to maximize the antitumor effects while minimizing side effects. The concept of tumor vascular normalization, which appears to be dose-dependent, suggests that lower doses of anlotinib may be preferable in certain combination regimens to improve drug delivery and reduce adverse reactions. Exploiting this window of normalization could be key to achieving long-term disease control.
• Exploration of Novel Indications:
Given its multi-targeted action and favorable pharmacokinetics, ongoing research continues to expand the potential indications for anlotinib. Studies in cancers such as medullary thyroid carcinoma, oral squamous cell carcinoma, and even osteosarcoma indicate that the drug’s benefit may extend well beyond NSCLC. Continued clinical trials in diverse tumor types will help establish its role as a cornerstone in targeted cancer therapy.
In summary, anlotinib Dihydrochloride’s mechanism of action is characterized by its potent inhibition of multiple receptor tyrosine kinases—most notably, VEGFRs, FGFRs, PDGFRs, and c-Kit—which disrupts both tumor cell proliferation and angiogenesis. At the molecular level, it blocks the activation of downstream signaling pathways such as PI3K/AKT and MEK/ERK, leading to impairment of cell survival signals and induction of cell cycle arrest and apoptosis. The drug’s capacity to normalize tumor vasculature further enhances its antiangiogenic profile and improves the efficacy of combination therapies. Clinically, these effects translate into prolonged progression-free and overall survival in a variety of cancer types including NSCLC, soft tissue sarcoma, hepatocellular cancer, colorectal cancer, and ovarian cancer. Despite its promising efficacy, challenges remain in the form of emerging resistance due to tumor adaptation, genetic variations, and microenvironment changes. Ongoing research aims to identify predictive biomarkers, optimize combination strategies, and refine dosing regimens to extend the clinical benefits of anlotinib.
Overall, anlotinib Dihydrochloride represents a significant advancement in the targeted treatment of cancer. Its general mechanism of blocking multiple key receptors provides a broad attack on the various hallmarks of cancer—from unchecked cell proliferation to angiogenesis—while its specific molecular actions allow for tailored combination therapies that can enhance treatment outcomes. Future directions in anlotinib research focus on overcoming resistance, identifying predictive biomarkers, and exploring novel therapeutic combinations, all of which are critical to maximizing patient benefit and advancing the field of precision oncology.
Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.
▶ See how 50+ research teams saved 300+ hours/month
From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.