What diseases does Trastuzumab treat?

7 March 2025
Overview of Trastuzumab
Trastuzumab is a recombinant humanized monoclonal antibody that specifically targets the extracellular domain of the human epidermal growth factor receptor 2 (HER2). Its mechanism of action principally involves binding to HER2 and subsequently inhibiting downstream signal transduction pathways that promote cell proliferation and survival. Additionally, trastuzumab mediates antibody‐dependent cellular cytotoxicity (ADCC), which helps the immune system recognize and eliminate HER2-overexpressing tumor cells. Overall, its ability to interfere with the cancer‐promoting effects of HER2 has revolutionized the treatment paradigm for patients with HER2-positive malignancies.

Mechanism of Action
Trastuzumab works by selectively binding to the extracellular domain of the HER2 receptor, thereby preventing receptor dimerization that is essential for activating downstream intracellular signaling pathways such as the PI3K/Akt and MAPK pathways. Its actions also involve the activation of immune effector cells via ADCC, leading to the targeted destruction of tumor cells. The binding of trastuzumab to HER2 not only blocks proliferative signals but also can induce receptor internalization and degradation, resulting in reduced receptor density on the tumor cell surface. This dual mechanism—both direct anti-proliferative effect and immune-mediated cytotoxicity—contributes to its robust antitumor activity.

Approval History
Trastuzumab was first approved by regulatory authorities in the United States in 1998 for the treatment of metastatic breast cancer in patients with HER2-positive tumors. Since its debut, trastuzumab has rapidly become a cornerstone in the management of HER2-driven cancers. Over time, its indications have been expanded to include early-stage adjuvant settings and metastatic disease scenarios, and a variety of biosimilars have been developed to improve accessibility around the globe. The continued evolution of its regulatory status reflects both its efficacy and importance in oncology.

Diseases Treated by Trastuzumab
Trastuzumab’s clinical utility is most prominently recognized in the treatment of cancers that overexpress the HER2 protein. Its use is well established in the management of HER2-positive breast cancer and HER2-positive gastric cancer. The precise targeting of HER2 defines the therapeutic landscape and makes the drug effective in specific tumor subtypes where HER2 signaling plays a pivotal role.

HER2-positive Breast Cancer
Among the diseases treated by trastuzumab, HER2-positive breast cancer is its foremost indication. Approximately 15–20% of breast cancers overexpress the HER2 receptor, and these tumors are known for their aggressive behavior and poorer prognosis when compared with HER2-negative counterparts.

In patients with early-stage, operable breast cancer, trastuzumab is administered in combination with chemotherapy in the adjuvant setting, where it has been shown to significantly reduce the risk of recurrence and improve overall survival. Multiple clinical trials have documented that incorporating trastuzumab into the standard adjuvant chemotherapy regimen results in durable benefits. In metastatic breast cancer, trastuzumab remains the standard of care either as monotherapy or in combination with chemotherapeutic agents, providing improved progression-free survival (PFS) and overall survival (OS) in patients with HER2-positive tumors.

Additionally, trastuzumab is frequently combined with other HER2-targeted therapies such as pertuzumab. This dual antibody approach has been observed to yield synergistic effects by blocking different epitopes on the HER2 receptor and thereby further interfering with HER2 signaling. Clinical trials employing such combinations have demonstrated not only improved efficacy but also the potential to delay disease progression and manage brain metastases in HER2-positive metastatic breast cancer. The introduction of antibody-drug conjugates (ADCs) like trastuzumab emtansine (T-DM1) has further expanded the therapeutic arsenal for HER2-positive breast cancer, enabling targeted delivery of cytotoxic agents with decreased systemic toxicities.

HER2-positive Gastric Cancer
Trastuzumab has also significantly impacted the treatment of HER2-positive gastric cancer and gastroesophageal junction (GEJ) adenocarcinomas. Gastric cancers tend to overexpress HER2 in a subset of patients, and the identification of this molecular phenotype has led to the delineation of “HER2-positive gastric cancer” as a distinct clinical entity.

Clinical trials, such as the ToGA trial, have demonstrated that the addition of trastuzumab to conventional chemotherapy (e.g., cisplatin plus capecitabine or 5-fluorouracil) significantly improves median overall survival and progression-free survival in patients with HER2-positive advanced gastric cancer. This benefit has been noted regardless of the chemotherapy backbone used, suggesting a robust effect linked primarily to HER2 antagonism. Trastuzumab achieves this by binding to the overexpressed HER2 receptors on gastric tumor cells, thereby interfering with HER2-mediated proliferation and triggering immune-mediated lysis.

While the initial indications were for advanced or metastatic disease, the success of trastuzumab in this context has spurred further research into its applicability in earlier lines of therapy and potentially in combination with novel agents such as immune checkpoint inhibitors and other targeted therapies. The repurposing of trastuzumab’s mechanism to treat gastric cancer reflects its versatility in addressing HER2-dependent oncogenic processes beyond breast cancer.

Clinical Efficacy and Outcomes
The efficacy of trastuzumab in improving survival outcomes in both breast and gastric cancers has been well documented through multiple clinical trials and real-world observational studies. Its ability to alter the natural history of HER2-positive malignancies is evident from the substantial gains in progression-free survival and overall survival reported in pivotal studies.

Clinical Trial Results
Large-scale clinical trials have consistently shown that trastuzumab significantly reduces disease recurrence and improves survival outcomes. In early-stage HER2-positive breast cancer, adjuvant trials have reported an absolute reduction in the recurrence risk and a marked improvement in long-term overall survival when trastuzumab is combined with chemotherapy. For instance, the NSABP B-31 and N9831 trials demonstrated that the addition of trastuzumab to standard chemotherapy resulted in a significant increase in disease-free survival, owing largely to a reduction in local and distant recurrences.

Similarly, in advanced or metastatic HER2-positive breast cancer, trials have documented that trastuzumab, either as a standalone treatment or in combination with other chemotherapies, leads to improved overall survival compared to regimens not including HER2-targeted therapy. In the realm of gastric cancer, the ToGA trial remains a landmark study, showing that the combination of trastuzumab with chemotherapy extended median overall survival compared to chemotherapy alone. These results validate the specific targeting of HER2 as an effective strategy in both breast and gastric malignancies.

The data derived from these trials are further supported by real-world evidence. Long-term safety analyses and observational studies have reiterated that while trastuzumab’s efficacy in controlling tumor progression is considerable, continuous monitoring and prompt management of adverse effects are necessary to ensure optimal patient outcomes.

Comparative Efficacy
When compared to traditional chemotherapeutic regimens in HER2-positive cancers, trastuzumab offers distinct advantages. Its targeted mechanism of action results in improved clinical outcomes with a favorable balance between efficacy and toxicity. By directly interfering with HER2 signaling, trastuzumab helps achieve higher response rates and greater rates of complete response in the neoadjuvant and metastatic settings.

Comparisons have been drawn between different administration strategies, such as intravenous versus subcutaneous formulations of trastuzumab. Studies have shown that subcutaneous formulations offer similar efficacy to intravenous infusions but with added benefits in terms of reduced patient chair time and improved convenience. Moreover, the development of biosimilars with equivalent efficacy profiles has paved the way for more cost-effective treatment options without compromising clinical outcomes.

Trastuzumab’s ability to enhance the effect of standard chemotherapy has also been compared favorably against sequential or monotherapy approaches in multiple studies. Notably, combination regimens that incorporate trastuzumab have demonstrated a significant synergy that translates to better therapeutic outcomes, especially in the first-line treatment of metastatic disease.

Safety and Side Effects
Despite its remarkable efficacy, trastuzumab is associated with certain side effects that require careful patient selection and vigilant monitoring throughout the treatment course. The safety profile of trastuzumab has been extensively characterized in numerous clinical trials, with cardiotoxicity emerging as the most significant concern.

Common Side Effects
The most frequently reported side effects of trastuzumab include infusion-related reactions and mild to moderate symptoms such as fever, nausea, and diarrhea. Acute infusion reactions tend to be manageable with appropriate premedication and supportive care, allowing most patients to continue therapy without significant interruption.

In addition, some patients experience flu-like symptoms, including asthenia and chills, during trastuzumab infusions. These adverse reactions are generally transient and are often mitigated by adjusting the rate of infusion or through prophylactic measures. The frequency of these side effects has been well documented, and they are typically considered acceptable given the overall survival benefits that trastuzumab confers in HER2-positive cancers.

Long-term Safety Profile
Cardiotoxicity is the primary long-term safety concern associated with trastuzumab therapy. Studies have shown that trastuzumab may lead to a decrease in left ventricular ejection fraction (LVEF) and occasionally symptomatic congestive heart failure, particularly when administered concurrently with other cardiotoxic agents such as anthracyclines. The risk of cardiotoxicity is dose-independent and can occur at any point during treatment, necessitating periodic monitoring of cardiac function via echocardiography or other imaging modalities every three months.

Long-term follow-up studies have demonstrated that while the majority of trastuzumab-induced cardiac effects are reversible upon treatment discontinuation and the initiation of heart failure management, a notable proportion of patients may experience persistent or late-onset cardiac dysfunction. This underscores the importance of careful risk stratification and cardiac assessment prior to and during therapy.

In addition to cardiotoxicity, rare but serious adverse events such as interstitial pneumonitis have been reported. Although these events are uncommon, they illustrate the need for comprehensive patient monitoring and prompt intervention in case of severe adverse reactions. Taken together, the safety profile of trastuzumab is favorable when balanced against its substantial clinical benefits, but the potential for adverse effects demands a multidisciplinary approach involving oncologists, cardiologists, and supportive care teams.

Future Research and Developments
The future of trastuzumab and its related therapies is focused on expanding its applications, optimizing treatment regimens, and developing novel combination therapies that can further improve clinical outcomes while minimizing toxicity.

Emerging Therapeutic Applications
Research is actively exploring new indications and earlier use of trastuzumab beyond the current standard-of-care settings. For instance, there is ongoing investigation into its use in the perioperative and adjuvant settings for HER2-positive breast cancer, with the goal of reducing recurrence rates even further.

Moreover, emerging clinical evidence suggests that trastuzumab might be effective in other HER2-positive malignancies. There are studies examining its efficacy in HER2-positive colorectal cancer, biliary tract cancer, and even leptomeningeal disease where HER2 signaling could be driving oncogenesis. While these applications remain investigational, early data indicate that the targeted disruption of HER2 may have beneficial effects in a broader range of tumors than previously recognized.

Researchers are also focusing on refining patient selection criteria by integrating advanced molecular diagnostics, such as circulating tumor DNA (ctDNA) and plasma tumor DNA assays. These tools aim to identify patients who are most likely to respond to HER2-targeted therapy, thereby personalizing treatment and potentially expanding the use of trastuzumab in patients with low or heterogeneous HER2 expression. The concept of “HER2-low” breast cancer is gaining traction, and studies are underway to ascertain whether patients with lower levels of HER2 expression might still benefit from trastuzumab-based therapies.

Novel Combination Therapies
Trastuzumab is frequently employed in combination with other therapeutic agents to achieve a synergistic antitumor effect. One of the most promising areas of research is the combination of trastuzumab with pertuzumab, another anti-HER2 antibody that binds to a different epitope of the HER2 receptor. This dual-targeting approach has been shown to provide enhanced blockade of HER2 signaling, leading to improved outcomes in both early-stage and metastatic settings.

In addition, the development of antibody-drug conjugates (ADCs) such as trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) represents a significant advance in the targeted delivery of cytotoxic agents. These novel agents combine the specificity of trastuzumab with the potent cell-killing ability of cytotoxic drugs, offering a more effective strategy for attacking HER2-overexpressing tumors while sparing normal tissue.

Furthermore, combination therapies involving trastuzumab and tyrosine kinase inhibitors (TKIs) such as tucatinib have demonstrated promising results in clinical trials, particularly for patients with brain metastases stemming from HER2-positive metastatic breast cancer. The complementary mechanisms of action—monoclonal antibody targeting versus small-molecule inhibition—permit more comprehensive suppression of HER2 signaling pathways and may overcome resistance associated with single-agent therapies.

Novel combinations are also being investigated with immunotherapeutic agents such as pembrolizumab, with the rationale that trastuzumab-mediated ADCC might synergize with immune checkpoint inhibitors to further augment antitumor immunity. Early-phase trials have explored these combinations in both breast and gastric cancer settings, with the goal of enhancing response rates and prolonging patient survival.

Conclusion
Trastuzumab has fundamentally transformed the management of HER2-positive malignancies over the past two decades. Initially approved for metastatic HER2-positive breast cancer in 1998, its efficacy was later established in the adjuvant setting for early-stage breast cancer, where it significantly reduces recurrence rates and improves overall survival. Moreover, the successful incorporation of trastuzumab into the treatment protocols for HER2-positive gastric cancer—validated by landmark trials such as ToGA—has expanded its therapeutic reach.

From a mechanistic perspective, trastuzumab exerts its antitumor effects by binding to the HER2 receptor and interfering with proliferative and survival pathways, while also recruiting immune effector cells through ADCC. This targeted action not only halts tumor progression but also enhances the effectiveness of other chemotherapeutic or targeted agents when used in combination.

While its clinical efficacy has been well established in both metastatic and early-stage settings, the safety profile of trastuzumab demands careful attention. Cardiotoxicity remains the most significant adverse effect, particularly when the agent is used in combination with other cardiotoxic drugs such as anthracyclines. Despite this, the overall tolerability of trastuzumab is acceptable, and ongoing research into optimal monitoring strategies and combination regimens continues to improve patient outcomes.

Looking ahead, future research is focused on expanding the indications for trastuzumab, refining patient selection through modern diagnostic techniques, and exploring novel therapeutic combinations. The integration of trastuzumab with other HER2-targeted therapies, ADCs, tyrosine kinase inhibitors, and even immunotherapeutic agents represents a promising avenue for overcoming resistance and enhancing treatment efficacy.

In summary, trastuzumab primarily treats HER2-positive breast cancer—in both early and metastatic stages—and HER2-positive gastric cancer, with emerging evidence suggesting potential applications in other HER2-dependent malignancies. Its evolution from a breakthrough monoclonal antibody to a multifaceted therapeutic agent underscores its central role in modern oncology. As research continues to refine and expand its applications, trastuzumab remains a vital tool in the fight against cancer, offering hope for improved survival and quality of life for patients worldwide.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

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.