Personalized Medicines: Disruptive Technologies in Life Sciences | Corning

The following article originally appeared on December 9, 2020 in Pharma's Almanac here.

Personalized treatments are poised to have a tremendous positive impact for patients across disease indications. However, a number of challenges to the discovery, development, manufacture, and administration of these novel therapies remain unresolved. Corning Life Sciences — through its many optimized products for organoid growth and creative tools and technologies for simplifying workflows, such as cell separation and engineering, among other activities — is committed to enabling the accelerated development and commercialization of precision medicines.

Pivotal Time Point for Personalized Medicines

The trajectory of personalized medicine is at a pivotal moment, owing to our expanding understanding of human biology and disease behavior and access to dynamic tools that continually increase our knowledge. More than ever before, we are able to visualize and analyze human physiological processes, gathering data in vivo, ex vivo, and in vitro using advanced imaging technologies and discovering previously unrealized patterns and connections using artificial intelligence and other computational tools.

In addition, technologies such as the expanding clustered regularly interspaced short palindromic repeats (CRISPR) toolbox and new mRNA approaches afford the ability to engineer genetic information. New models that mimic complex human anatomy in a dish or on a chip via tissue engineering are disrupting conventional drug discovery models. In particular, chimeric antigen receptor (CAR) — patient T cells genetically engineered to express antigen receptors targeting cancer cells — and organoid technologies (tiny, self-organized three-dimensional tissue cultures derived from stem cells) will continue to disrupt the cell and gene therapy and regenerative medicine fields, translating early successes with liquid cancers to solid tumors and beyond. 

Overall, these technologies will continue to drive the further development of personalized medicine, with all of them ultimately working together to match the right patient with the right treatment at the right time. One challenge to overcome, though, will be the higher cost of these novel solutions. Once their effectiveness is definitively realized, the focus will shift to making them more efficient and accessible. In addition, once it has been demonstrated that these technologies have broad usability, they will serve as a foundation that can help reduce risks — both to patients and in terms of costs and timelines — of future clinical trials.

Organoids and Precision Medicine

Organoids present enormous potential in both drug development and precision medicine. When effectively leveraged, they can reduce the cost and risks associated with novel therapies by enabling precisely targeted treatments for individual patients. With their ability to serve as valuable model systems for studying the tumor microenvironment, organoids are accelerating discovery efforts in cancer biology and enabling rapid screening of potential new therapeutics. 

Organoids are generated using tissue-specific stem cells, which are typically undifferentiated epithelial cells. It is also possible to generate organoids from patient cells to investigate the genetic alterations within an individual’s cancer cells. Of particular interest is the ability to culture precancerous cells that are not fully transformed, which have not previously been accessible to the research community.

For instance, tumor organoids in a dish can be generated from cells collected during a biopsy by mixing them with a semisolid matrix and then exposing the mixture to media containing growth factors. The resultant organoid can be used for drug screening rather than administering different medications to the patient to see which might provide a positive response. These types of organoids can also be inserted into mice (xenografts) that can be challenged with different therapeutics. 

In some cases, organoids provide a means for evaluating potential therapies where no practical solution currently exists. At present, postmortem human retinal explants are the only human models available that achieve a reasonable level of complexity for the evaluation of ophthalmology treatments. Results from small animal models are often not translatable to humans, as they do not fully represent the human retinal system. Organoids thus offer the potential to transform the paradigm for drug discovery and development for eye diseases. 

As part of the effort to develop a treatment for COVID-19 infections, organoids are being used to determine the mechanistic pathways by which the novel SARS-CoV-2 virus affects different organs in the body, such as the lungs, liver, and kidneys.

Moving Beyond the Basics

While organoid technology has been continuously evolving, challenges remain, particularly with respect to the creation of vascularized, multilineage organoids containing the blood vessels needed to provide oxygen and nutrients, remove metabolic waste, and facilitate communication between different cell types. All of these capabilities are essential for them to mature into fully functional tissue building blocks. Technology improvement is needed that can overcome this limitation and allow for the integration of organoids into clinical practice. Recently evolved microfluidic technology, which includes a variety of 3D fabrication techniques, presents one opportunity for achieving this goal. 

More progress will also be needed on the scale-up of organoids to facilitate their use in the development of new drugs and personalized treatments and to increase their potential to reduce the number of clinical trials required. Effective validation of organoids to ensure that they accurately recreate the tissue of interest must also be consistently achieved. In addition, because purity is crucial, solutions are needed to ensure that contaminants and undesired cell types are removed from organoids derived from tissues.

Finally, while organoids have been developed that represent organs such as the heart, liver, and kidneys, scientists are looking to expand into new models that possess more functionality. One primary example is pulmonary organoids that can replicate the air–liquid interface, which would be valuable for both fundamental research and drug testing.

From Organoids to Engineered Tissues

Creating engineered tissues with more complex functionality is the next goal in the organoid field. Recent developments in cell culture technology have opened up new opportunities for improved physiological cell-based assays for disease models and regenerative medicine. By combining fabrication and 3D printing technology with cellular biology, for example, it is possible to create 3D printed organoids that better mimic in vivo conditions. 

However, the challenge remains to ensure that the engineered tissue has vasculature, the correct composition of different cell types (i.e., neuronal cells/fibers, cardiomyocytes, skeletal myocytes, smooth muscle cells, etc.), and the appropriate ratio of different types of innervation (sympathetic, parasympathetic, sensory, and enteric phenotypes). 

True engineering of organs-in-a-dish, therefore, will require extensive collaboration between different functional areas, such as developmental biology, stem cell biology, biomaterials, 3D biofabrication, and regenerative medicine.

Some progress has been made with organoids and organ-on-a-chip systems. In one case, liver, heart, and lung organoids were bioprinted to develop a multiorgan-on-a-chip platform for the investigation of the interactions between organs and their individual and collective responses towards drugs and toxins.1

Onto Biofabrication for Transplantation

Given the significant shortage of donated organs, the ultimate goal for many organoid developers is the biofabrication of full organs suitable for transplant into humans. Here again, creating vasculature is the big hurdle. 

The development of vasculature does not involve endothelial cells alone. Other cell types, such as pericytes, smooth muscle cells, and immune cells, play roles in providing structural and functional support, as well as signaling guidance. The development of technologies that enable the incorporation of these multiple cell types will be required to build more comprehensive in vitro models. 

With their ability to serve as valuable model systems for studying the tumor microenvironment, organoids are accelerating discovery efforts in cancer biology and enabling rapid screening of potential new therapeutics.

As a start, 3D microfabrication technologies are helping organoid developers overcome the lack of vascularization in organoid model systems. Indeed, a variety of bioengineering approaches, such as organ-on-chip and 3D printing technology, can establish 3D microenvironments for organoids that mimic the physiological environment. In addition, microvascular patterning and microfluidic technology will allow the incorporation of the right cells at the right locations. Furthermore, human stem cells — with their high potency and ability to proliferate and differentiate into multiple cell types — are well suited for organ fabrication. 

To date, some organoids have been transplanted in animal models for investigation of their behavior. In one study, a method was developed for successful transplantation of human brain organoids into the adult mouse brain.2 The organoid grafts were observed to progressively differentiate and mature, resulting in functional neuronal networks interconnected synaptically with host neuronal circuits and extensive infiltration of the host vasculature within a few days after transplantation.

In another example, kidney organoids transplanted into a mouse model of unilateral urethral obstruction (UUO) were observed to become vascularized and found to survive in the transplanted graft for at least two weeks after UUO and transplantation.3 Organoids are also being explored for the treatment of liver diseases. Bioengineering approaches for liver cell therapy using liver spheroids/organoids as transplantable units may provide suitable alternatives to further improve liver regeneration and therapy.

Facilitating the Organoid Field

Work at Corning Life Sciences is helping the organoid field to expand and achieve its full potential. Our goal is to provide the research community with better tools and resources for organoid applications and to further the science of organoid models by listening to their needs and translating them into products that add value. 

The extracellular matrix (ECM) is an important component in generating 3D models, because it provides biochemical properties and structural support that helps mediate signaling for cell migration, cell behavior, and polarization in organoid structures.

Corning provides the gold standard ECM — Matrigel® matrix — for organoid research and disease modeling. We recently launched a new version of the Matrigel matrix for organoid culture. This formulation has been optimized to support growth and differentiation of organoids from both healthy and diseased cell origins. Each lot of Matrigel matrix for organoid culture has been measured for matrix stiffness (elastic modulus) to best support organoid workflow. Each lot is also pre-qualified to form stable “3D dome” structures commonly used in organoid culture. As a result, the reproducibility and consistency that is essential for organoid research is achieved while reducing the need for time-consuming screening. 

The throughput for screening with 3D models has also been increased by Corning with the launch of high-throughput plate formats that provide ready-to-use options for drug screening with organoids. The Corning Matrigel matrix-3D  96- and 384-well microplates come pre-dispensed with Matrigel matrix and provide the reproducibility and consistency required for drug discovery research resulting in saving time. Work can be further accelerated by leveraging the range of Application Notes prepared by Corning scientists on the use of our products for organoid development.

Corning also provides media, growth factors and plastic consumables unique to the market and optimized for organoid culture environments, including spheroid microplates that enable 3D growth based on many different cell types. Via collaborations with the Hubrecht Organoid Technology (HUB), a pioneer institute that amplifies the work of Professor Hans Clevers and his methods for growing stem cell–derived human “mini-organs” (HUB Organoids), Corning has demonstrated applications supporting the culture of various organoids using Corning Matrigel matrix and our other consumables. 

Improving Workflows for Other Personalized Medicines

Corning Life Sciences is not only interested in facilitating the development and application of organoids for precision medicine. Cell and gene therapy and immunotherapy are other major areas of focus. Immunotherapy refers to any therapies that work by activating or suppressing a patient’s immune system. While the field includes more conventional immunomodulator therapies, including interleukins, cytokines (such as interferon), and immunomodulatory imide drugs, the focus in immunotherapy for cancers (immuno-oncology) has been on CAR-T therapies. In these modalities, T cells are harvested from a patient (autologous therapies) or a healthy donor (allogeneic therapies) and genetically engineered to express a particular CAR selected to target an antigen present on the surface of tumors. Once these CAR-T cells encounter a tumor antigen, they activate, proliferate, and become cytotoxic, leading to destruction of the tumor cells. Following FDA approval of Novartis’ Kymriah and Kite Pharma’s Yescarta, we have seen an explosion of clinical trials exploring CAR-T therapies for blood cancers and solid tumors. However, safety concerns surrounding long-term effects — particularly concerning cytokine release syndrome and neurological toxicity — remain. CAR design has already undergone a number of generations of design, adding co-stimulatory domains and cytokines and synthetic control mechanisms to increase efficacy. Despite growing efficacy data, challenges remain to manufacturing these therapies, including the complexity of their manufacturing and the needed supply chain infrastructure, a logistically challenging, high-touch commercial model, and the high costs and ensuing reimbursement challenges.

Our goal is to provide the research community with better tools and resources for organoid applications and to further the science of organoid models by listening to their needs and translating them into products that add value

We understand that developing and manufacturing high-quality clinical-grade cell therapies for large-scale patient use remains a challenge. To bring ground-breaking treatments to patients as quickly as possible, researchers and manufacturers need to improve on manual, time-consuming processes. 

Corning has many roles to play in facilitating cell therapy workflows, from the time blood samples are collected through separation of blood components, viral vector manufacture, cell engineering, expansion, and formulation. 

Recently, we launched the X-SERIES cell-separation platform to help streamline and automate many of the steps involved in cell processing. This flexible, multisystem platform provides a streamlined solution and performance gains in purifying mononuclear cells, washing contaminants from cell fractions, selecting specific cell populations, and concentrating cellular suspensions. All are crucial steps in the development of life-changing cell therapies.

Beyond enabling these activities, we are also supporting researchers in their creation of organoid models that closely resemble patient tumors, both across patient populations and on the individual level, that aid in the development of treatments with higher efficacy with reduced risk.

While immunotherapy and cell and gene therapy are in their infancy, they are growing rapidly, and advances are bringing the possibility of developing effective treatments for solid tumors. Facilitating the development of such life-changing therapies and making them accessible and affordable for all patients is a great honor that also brings great responsibility.

We take that responsibility seriously and are committed to creating and collaborating on new tools that enable further technological advances and developing solutions that simplify and optimize workflows for researchers to develop personalized medicines. 

References

  1. Skardal, Alexander, et al. “Multi-tissue interactions in an integrated three-tissue organ-on-a-chip platform” Sci. Rep. 7:8837 (2017).
  2. Mansour, Abed, et al. “An in vivo model of functional and vascularized human brain organoids” Nat Biotechnol. 36:432–441 (2018).
  3. Nam, Sun Ah, et al. “Graft immaturity and safety concerns in transplanted human kidney organoids.” Experimental & Molecular Medicine. 51:145 (2019).