Big Pharma Mega Deals in the Last Few Months Series Part 2 | Next-Generation Oncology: in vivo CAR-T, ADCs, Bispecifics and Oral Biologics

Among the many big pharma transactions announced in the last few months, oncology stands out as the area where activity is both dense and conceptually rich. Not only are we seeing large deals in terms of headline value, but they are also tightly linked to a new wave of modalities: in vivo CAR-T, antibody-drug conjugates (ADCs) including bispecific formats, and oral biologics.

This article is Part 2 of the “Big Pharma Mega Deals in the Last Few Months” series and focuses on oncology. Building on the overview in Part 1, we zoom in on how recent deals illustrate the evolution from classic antibodies and small molecules toward a layered landscape of advanced modalities.


TOC

1. Why start the deep dive with oncology?

1-1. Still the most critical frontier for large pharma

For more than a decade, oncology has been the single most important growth driver for many large pharmaceutical companies. Immune checkpoint inhibitors, targeted therapies, and CAR-T cell therapies have created blockbuster franchises, and pipelines remain heavily weighted toward cancer indications.

Yet, the unmet need in oncology is still substantial. Resistance, relapse, and the complexity of tumor biology mean that even in indications with multiple approved agents, there is plenty of room for improvement. This depth and breadth of opportunity continue to justify sustained, large-scale investment in oncology R&D and deal-making.

1-2. The most intense arena of modality competition

Oncology is not only rich in targets and indications; it is also the most competitive arena in terms of modalities. In the last few months of deals, we see:

  • in vivo CAR-T, aiming to re-engineer T cells inside the body
  • ADCs and bispecific antibodies, designed to improve selectivity and potency
  • Oral biologics, which have the potential to transform how therapies are delivered

These new modalities sit on top of existing layers like monoclonal antibodies and small molecules, turning oncology into a multi-layered ecosystem rather than a single-modality battlefield.


2. Recent oncology deals that capture this shift

In this article, we will keep the following transactions in mind as emblematic of how big pharma is repositioning itself in oncology:

  • AbbVie – Capstan Therapeutics: a major move into in vivo CAR-T and related in-body cell engineering platforms
  • Takeda – Innovent: a large oncology collaboration built around a PD-1/IL-2α-bias bispecific backbone (IBI363) and a CLDN18.2-targeted ADC (IBI343)
  • Chugai – Rani Therapeutics: a multi-program partnership using the RaniPill® oral delivery platform for biologics, with oncology-relevant implications

Each of these deals highlights a different angle: scalability of cell therapies, next-generation antibody formats, and transformation of delivery routes.


3. in vivo CAR-T: the next stage of cell therapy

3-1. From ex vivo CAR-T limitations to in vivo concepts

Traditional CAR-T therapies are largely “ex vivo”: T cells are harvested from the patient, engineered and expanded in a manufacturing facility, and then reinfused. While this approach can deliver dramatic clinical benefits, it comes with well-known challenges:

  • Highly complex and expensive manufacturing processes
  • Limited scalability, as each product is bespoke
  • Dependence on specialized facilities and skilled personnel

in vivo CAR-T aims to sidestep these constraints by engineering T cells directly inside the body. Using delivery vehicles such as nanoparticles or viral vectors, in vivo approaches seek to:

  • Reduce manufacturing cost and lead time
  • Make cell therapies accessible to a broader patient population
  • Extend the concept beyond oncology into autoimmune and other diseases

3-2. Strategic significance of the AbbVie–Capstan deal

The AbbVie–Capstan transaction is best understood as a platform acquisition rather than a single-product bet. Key aspects include:

  • A focus on a technology that can be applied across multiple indications, not just one asset
  • The potential to address both oncology and non-oncology immune-mediated diseases
  • A recognition that the core architecture of cell therapy manufacturing may need to be redesigned

In other words, this is an attempt not to patch the current ex vivo paradigm, but to explore a fundamentally different configuration of cell therapy.

3-3. Potential industry impact if in vivo CAR-T succeeds

If in vivo CAR-T proves to be clinically effective and commercially viable, the impact could be profound:

  • Moving from patient-specific manufacturing toward more scalable, batch-like processes
  • Expanding the number of patients and indications that can realistically be treated with cell-based approaches
  • Requiring new frameworks to address off-target effects, long-term safety, and regulatory considerations around in-body gene manipulation

The recent mega deal signals that at least some big pharma players see this as a risk worth taking at scale.


4. Bispecifics and ADCs: PD-1/IL-2α and CLDN18.2 as examples

4-1. Takeda–Innovent: combining a backbone and a target

The Takeda–Innovent collaboration brings together a PD-1/IL-2α-bias bispecific antibody (IBI363) and a CLDN18.2-targeted ADC (IBI343) in a single strategic package. Conceptually, this can be thought of as combining:

  • A backbone that modulates T-cell responses (PD-1/IL-2α)
  • A highly tumor-selective weapon (CLDN18.2 ADC) for specific cancer types

The bispecific backbone can enhance immune activation, while the ADC delivers potent payloads to tumor cells expressing CLDN18.2. Together, they reflect a layered approach to improving efficacy and selectivity.

4-2. Where the value of bispecific antibodies lies

Bispecific antibodies are not simply “two targets in one molecule.” Their value lies in the ability to:

  • Control the spatial and temporal relationship between targets at the molecular level
  • Design specific mechanisms of action, such as T-cell recruiting or signal modulation
  • Embed combination logic into a single agent, potentially simplifying treatment regimens

A PD-1/IL-2α combination, for example, can be seen as a way to tune the “accelerator and brakes” of the immune system within one programmable construct.

4-3. CLDN18.2 ADC and the idea of “organ-specific” cancers

CLDN18.2 is highly expressed in certain gastric and pancreatic cancers, making it a compelling target for ADCs. In a sense, CLDN18.2-targeted ADCs turn “organ-specific” cancers into antigen-defined subsets that can be treated with more precise payload delivery.

  • High local drug concentration at the tumor site
  • Opportunities to vary payloads to address resistance and toxicity profiles

The Takeda–Innovent deal can therefore be read as an effort to secure a position in a growing wave of antigen-driven ADC strategies in solid tumors, particularly in gastrointestinal oncology.


5. Oral biologics: what changes when the route of administration changes?

5-1. Chugai–Rani: the RaniPill® platform in context

The Chugai–Rani partnership may initially look like a niche play—oral delivery of biologics for selected rare diseases. However, the underlying concept is broader:

  • Enabling oral administration of antibodies and peptides that are currently injectables
  • Improving patient adherence and quality of life
  • Potentially reshaping how outpatient and infusion resources are used

Platforms like RaniPill® are not oncology-specific, but their relevance to oncology is obvious when one considers long-term maintenance therapies, adjuvant settings, and combinations that require repeated dosing.

5-2. Why this matters for oncology

In oncology today, many treatments are administered as intravenous infusions in hospital or clinic settings. This creates:

  • Significant burden on patients in terms of time and travel
  • Resource constraints for healthcare systems and infusion centers
  • Challenges in sustaining treatment over long periods

If even a subset of biologics could be delivered orally, potential benefits would include:

  • Reduced visit frequency for patients
  • Reallocation of clinical resources toward more acute or complex care
  • Improved long-term adherence in maintenance or adjuvant therapy

Recent deals around oral biologics can thus be interpreted as early steps toward rethinking not just efficacy, but the entire care pathway in oncology and beyond.


6. Shared strategic themes across these three modalities

in vivo CAR-T, ADCs/bispecifics, and oral biologics may appear quite different technically, but they share several strategic themes.

6-1. Value is being defined beyond “how strong the drug is”

Historically, oncology innovation has focused strongly on efficacy metrics such as response rates and progression-free survival. These remain central, but the recent wave of deals suggests a broader definition of value that includes:

  • Manufacturing cost and scalability
  • Route and frequency of administration, and their impact on quality of life
  • Operational burden on healthcare systems

in vivo CAR-T targets the manufacturing and scalability problem; oral biologics aim at route and convenience; ADCs and bispecifics try to raise the therapeutic index by improving selectivity.

6-2. Oncology as a stack of layers rather than a single plane

Another way to think about these developments is to view oncology as a stack of layers rather than a set of isolated lines of therapy:

  • Checkpoint inhibitors as one layer
  • ADCs and bispecifics as an additional precision layer
  • Cell therapies as yet another layer for selected patients and settings
  • Oral biologics as a layer that reshapes chronic and maintenance treatment

Recent mega deals can be interpreted as attempts to secure “slots” in multiple layers of this stack, rather than staking everything on a single modality or mechanism.


7. How investors and BD teams can read oncology deals

Part 5 of this series will discuss deal interpretation in more detail, but even within oncology alone, a few lenses are useful for investors and BD professionals.

7-1. Is this primarily a product play or a platform play?

One key question is whether a deal is fundamentally about:

  • A single lead asset with well-defined near-term value
  • A platform that can generate multiple assets across several indications

in vivo CAR-T and oral biologic delivery platforms are classic examples of the latter; ADCs may sit somewhere in between, being both target- and payload-dependent.

7-2. Which layer of the oncology stack is being targeted?

It also helps to ask:

  • Is this modality meant to add a new layer on top of existing standards of care?
  • Is it aiming to replace an existing layer?
  • Or is it currently an experimental layer that might move toward the standard over time?

Positioning a deal within this layered view of oncology can clarify both its risk profile and its potential strategic importance.


8. Implications for researchers, clinicians, and startups

From the vantage point of the last few months of oncology deals, we can draw some tentative implications for different stakeholders:

  • For researchers: It becomes increasingly important to frame projects not only around a single target or pathway, but also around where the modality sits in the broader treatment stack and how it could be combined with other layers.
  • For clinicians: As treatment options proliferate, decisions will hinge not only on efficacy, but also on how realistic it is for patients to adhere to complex regimens over time.
  • For startups: Beyond having “cool science,” it is critical to articulate which gap in pharma’s oncology portfolio is being filled—or which entirely new layer is being created.

Seen this way, recent mega deals are not just external events, but useful reference points for refining one’s own research or business strategy.


9. My thoughts and future outlook

Looking across the recent oncology deals, it feels less and less useful to frame the landscape as a simple competition between modalities—CAR-T versus ADCs, bispecifics versus small molecules, and so on. Instead, what emerges is a picture of an expanding stack of therapeutic layers, each with its own strengths, limitations, and natural use cases. in vivo CAR-T, ADCs, bispecifics, and oral biologics do not simply displace one another; they are gradually finding distinct roles within a more complex treatment architecture that spans lines of therapy, patient subgroups, and patterns of use over time.

At the same time, this growing richness creates new challenges for everyone involved. For patients and clinicians, navigating a menu of increasingly sophisticated options can be overwhelming, especially when choices have to be made under time pressure or in the context of limited resources. For developers and investors, it becomes essential to anchor decisions not only in efficacy data, but also in realistic assumptions about how therapies will be delivered, combined, and maintained in real-world practice. My own sense is that the next few years in oncology will be shaped less by single “magic bullet” breakthroughs and more by the careful design of combinations, sequences, and care pathways that make intelligent use of this expanding toolbox. If this series can help readers think about oncology in that more layered and practical way, it will have served its role.

This article was prepared by the Morningglorysciences editorial team.

Related Article

Pharma & Biotech News

Biotech Investment

Comment Guideline

💬 Before leaving a comment, please review our [Comment Guidelines].

Let's share this post !

Author of this article

After completing graduate school, I studied at a Top tier research hospital in the U.S., where I was involved in the creation of treatments and therapeutics in earnest. I have worked for several major pharmaceutical companies, focusing on research, business, venture creation, and investment in the U.S. During this time, I also serve as a faculty member of graduate program at the university.

Comments

To comment

CAPTCHA


TOC