#Oncology#DrugDevelopment#Bioanalysis#Immunoassay#PrecisionMedicine#Pharma#Biotech#ADCs#ClinicalResearch

The Evolution of Cancer Therapy: How the 2025 Approval Wave Sets New Bioanalytical Standards

1. Oncology Innovations in 2025: A Wake-Up Call for Precision Analytics

The first half of 2025 marked a turning point in oncological drug development. The U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) gave the green light to a remarkable number of new therapeutic approaches that have the potential to fundamentally change cancer treatment. This wave of approvals is not only an indicator of the industry’s continued innovative strength but also a clear impetus for the bioanalytical sciences, which are now challenged to keep pace with this rapid development. The complexity of these new drug classes presents significant challenges to conventional analytical methods and makes the implementation of advanced technologies essential.

 

Ask the Scientists

1.1. A Look at FDA and EMA Decisions in the First Half of 2025

In the first quarter of 2025 alone, the FDA and EMA approved 39 new or expanded indications for previously approved drugs, along with four entirely new oncological therapeutics. These decisions included treatment options for a total of approximately 14 types of solid tumors and 8 hematological malignancies. A significant proportion of these approvals, roughly three-quarters, were for biologics or biosimilars, which underpins the industry’s strategic shift away from classic small molecules toward more complex biological therapeutics.

Among the most notable approvals are groundbreaking therapies that are pushing the boundaries of precision oncology. These include innovative antibody-drug conjugates (ADCs) such as Datopotamab deruxtecan and Trastuzumab deruxtecan, approved for the treatment of breast cancer. Bispecific antibodies like Linvoseltamab for multiple myeloma also received approval in Europe, highlighting the potential of this new class of immunotherapeutics. However, the advances are not limited to antibody-based approaches. For example, the FDA granted approval for a gene therapy called ZEVASKYN for the treatment of wounds in patients with recessive dystrophic epidermolysis bullosa, signaling the maturation of this highly specialized form of therapy.

This therapeutic innovation is directly and causally linked to a growing analytical need. Where traditional approaches could assess the efficacy and safety of small molecules, the new, complex modalities demand an equivalent level of analytical innovation. Conventional bioanalytical methods, optimized for less heterogeneous molecules, are now reaching their limits. The consequence of this development is that companies that want to remain competitive in this dynamic environment must rethink their analytical strategies and switch to the next generation of technologies to meet the new requirements.

Ask the Scientists

1.2. Strategic Shifts in the Cancer Therapy Pipeline

The approvals in the first half of 2025 reflect the strategic trends throughout the entire oncology pipeline. There is an increased focus on developing therapeutics for what were previously considered “undruggable” targets, as well as refining existing approaches. For example, bispecific antibodies are ushering in an “immuno-oncology renaissance” by specifically guiding T-cells to tumor cells, thereby triggering a highly specific immune response. These new molecules are being investigated against a wide range of targets such as HER2, CEA, EpCAM, and GD2.

In parallel, the development of antibody-drug conjugates (ADCs) is progressing at a rapid pace. The focus here is on identifying new targets and optimizing ADC design to increase effectiveness while minimizing systemic toxicity. Progress is also being made in cell and gene therapies, with ongoing developments in CAR-T cells and the approval of the first tumor-infiltrating lymphocyte (TIL) therapy for advanced melanoma. Another significant breakthrough is the targeted treatment of KRAS mutations, which have historically been considered extremely difficult targets to treat. The success with such “undruggable” targets and the focus on rare diseases show that personalized medicine is no longer a vision, but increasingly shaping clinical reality.

These shifts in the pipeline have an important consequence for bioanalytics: most of these innovative approaches are associated with extremely low drug concentrations or limited sample volumes from rare patient populations. Consequently, bioanalytical methods are needed that can precisely measure even these minimal amounts. Another connection is revealed in the increasing integration of technologies such as artificial intelligence (AI) and machine learning (ML) in cancer research. These technologies are used to identify predictive biomarkers and personalize treatment. It is important to note that AI models are based on reliable and accurate data. Without the high-sensitivity bioanalytical data obtained through advanced immunoassay platforms, these AI-driven approaches would, however, be largely useless.

Ask the Scientists

2. The Analytical Challenge: Why the Next Generation of Cancer Therapy is Pushing Bioanalytics to its Limits

The complexity of the new therapeutic approaches goes far beyond what is expected for conventional small molecules. This creates specific, multifaceted problems for bioanalysts and R&D leaders that fundamentally influence strategic decisions in drug development. The need to measure extremely low concentrations in minimal sample volumes while meeting the strictest regulatory requirements is not just a technical hurdle but a critical bottleneck in the entire development process.

2.1. The Strategic Importance of PK/PD Data in Drug Development

The precise collection of pharmacokinetic (PK) and pharmacodynamic (PD) data is the foundation of any successful drug development. This data is not merely a regulatory requirement but the basis for critical “go/no-go” decisions in the early clinical phases. A comprehensive understanding of PK profiles, which includes bioavailability, tissue exposure, and drug clearance, is crucial for determining the correct dose and optimal dosing schedule.

In parallel, pharmacodynamics is essential for assessing the biological effect of the drug candidate. Pharmacodynamic biomarkers provide evidence that the drug reaches its target and triggers the desired physiological or molecular response. These biomarkers are an indispensable tool for demonstrating efficacy and enabling an early assessment of the therapeutic response. Low-dose studies or microdosing are more economically efficient and less risky for patients, but they can only be conducted if the analytical method is sensitive enough to capture drug and biomarker concentrations in the femtogram-per-milliliter range. A technology that reliably measures these low concentrations enables informed decision-making, accelerates the development process, and reduces the risk of costly misinvestments.

Ask the Scientists

2.2. The Analytical Complexity of Antibody-Drug Conjugates (ADCs)

Antibody-drug conjugates (ADCs) are a prime example of the increased analytical complexity in modern oncology. Unlike conventional monoclonal antibodies, ADCs are not homogeneous molecules but highly diverse mixtures consisting of an antibody, a linker, and a cytotoxic payload. For sound bioanalysis, it is essential to differentiate between the various molecular species: the intact ADC, the conjugated payload, and the released, unconjugated payload.

The detection of the free payload is of crucial importance, as it can be responsible for systemic toxicity, while the concentration of the intact ADC determines therapeutic efficacy. The challenge becomes even more complex due to factors such as the instability of the ADC and payload in the sample matrix, potential ex vivo artifacts during sample collection that can lead to artificial payload release, and the inaccuracy of measuring low toxophor concentrations in the presence of a large excess of ADC. The heterogeneity of ADCs and the need to quantify multiple species simultaneously is one of the greatest technological limitations for laboratories that rely on conventional ELISA methods. These shortcomings are the main driver for the search for more advanced, specific, and sensitive assays that also have multiplexing capabilities to meet the growing regulatory and scientific requirements.

2.3. The New Frontiers: Ultra-Sensitivity and Microsampling

One of the greatest hurdles in the preclinical and clinical development of modern therapies is the need to measure extremely low concentrations of biomarkers or drugs in minimal sample volumes. High-sensitivity immunoassays are indispensable for this, as they make it possible to create complete PK/PD profiles that could not be captured with traditional methods. Detection in the femtogram/mL or picogram/mL range is necessary to track the effect of drugs in microdosing studies or in samples from rare matrices, such as cerebrospinal fluid (CSF) or intraocular fluid. These samples are often available only in very limited quantities, which is why the analytical platform must be able to work with sample volumes in the microliter range.

The need for microsampling and the detection of rare analytes has far-reaching consequences. It is not just a technical requirement but has a direct impact on ethical considerations and patient recruitment. By reducing sample volume, studies can be conducted with sensitive patient populations, such as children with brain tumors, or in areas with limited sample availability. This, in turn, increases the potential for developing therapies for rare diseases and unmet medical needs. The ability to robustly and reproducibly generate data from the lowest concentrations is therefore key to minimizing risks in drug development and achieving unparalleled data quality.

3. Technological Answers: How Next-Generation Immunoassay Platforms Master These Challenges

The rapid advances in oncological therapy require an equally fast development of bioanalytical tools. The market dynamics, driven by new biologics and stricter regulatory requirements, have exposed the limits of traditional immunoassay methods. Conventional ELISA platforms, which often do not achieve the necessary sensitivity to measure the low concentrations of modern drugs, are increasingly inadequate. This has created a demand for advanced technologies such as Immuno-PCR (Imperacer®), Simoa®, and MSD™ that are capable of meeting these challenges.

Ask the Scientists

Experience the power of biotechnology – learn more now!

Unlock the potential of advanced biotech.