Tumor Organoids Are The Future Of Cancer Research

Organoids are three‐dimensional (3D) miniaturized versions of organs or tissues derived from patient cells that mimic the essential function, structure, and biology of those tissues or organs. Tumor organoids, then, are 3D versions of a patient’s cancer cells that allow for exceptionally accurate ex vivo testing.

Why Not Stick With 2D Cultures?
Performing accurate studies on mammalian cells requires growing the cells in an environment that mimics the body as closely as possible. Researchers have been growing cells on flat surfaces (2 dimensions, or 2D), since the early 1900s. While 2D models play a vital role in research, they have several limitations in that tumors grow in 3D environments inside the body.

For 2D, the adhesion to the hard plastic surface inhibits the capacity for cells to form multi-dimensional structures. These conditions alter the morphology of the cells resulting in loss of diverse phenotype and unable to mimic the tumor microenvironment which is essential to stimulate the heterogeneity of cancer cells

With new techniques, researchers can now grow cells in a 3-dimensional space, usually embedding the cells within a gel-like matrix or growing in a solid scaffolding. The growth of these cells allows better cell-to-cell contact. A real advantage over 2D is that 3D mimics tissue architecture and cell-cell interactions more accurately.  Since 3D cultures can model a tissue while being cultured in vitro, this makes them ideal for research that focuses on morphology, proliferation, differentiation, response to stimuli, drug metabolism, and protein synthesis.

The 3D developing technique that’s paving the way are tumor organoids.

Remarkable Clinical Predictivity
Tumor organoids owe their remarkable clinical predictivity to their recapitulation of the key phenotypic and morphologic features of the original tumor. This allows for patient-derived tumor organoids to respond to chemotherapy in nearly the same way as cancer cells growing in the body would respond.  The adoption of being able to resemble the native organs in terms of gene and protein expression and microscale tissue architecture will help revolutionize and personalize therapeutic development.

Researchers can embed bits of the tumor organoids across several different wells then apply different treatments to each cell to determine which might give the patient the best possible therapeutic result.

Unlike cells grown in 2D, organoids grown in 3D environments retain the characteristics of the original tumor, its heterogeneity, and even predict response to treatment – all of which makes Patient Derived Organoids a model system for precision medicine and preclinical cancer research.

Cost-Effectiveness
Cost is always a consideration, of course. Compared with patient-derived xenografts (PDX), which is another in vivo model in which human patient tumor material is implanted into immunodeficient mice, organoids are less expensive, require less labor, and propagate faster. PDX requires costly long-term housing of mice, for example, whereas patient-derived organoids are embedded into a matrix that is easy and inexpensive to care for.

Provides An Opportunity To Research Patient-Derived Tumors At Various Stages of Carcinogenesis
Organoids provide researchers with unique opportunities to study patient-derived tumors. From tumor genesis to metastasis, for instance, cancer cells undergo multistage transformation towards carcinogenesis. The changes they undergo can affect how the cells respond to treatment – a drug that works well at tumor initiation may not be effective during malignant conversion, for instance.

Tumor cell-derived organoids may be collected at various stages of carcinogenesis, which can provide researchers with valuable insight into how their treatment modalities may have to change over time.

Organoids are also exceptional predictive biomarkers, which are critical for enabling personalized medicine, improving treatment efficacy, and increasing survival for cancer patients. Predictive biomarkers are essential for reducing toxic side effects and decreasing treatment costs. Patient Derived Organoids (PDOs) make individualized tumor response testing possible. A growing number of studies have examined PDOs since 2018 and have identified PDOs as a reliable predictive biomarker in cancer treatment.

A Growing Number of Researchers Are Moving Towards Patient Derived Tumor Organoids – And For Several Great Reasons
Ease of production makes it possible to make organoids from the same patient at several points in time or at various stages of tumor development, which gives researchers insight into how tumors change over time. Scientists may use organoids to gain a better understanding of exactly when certain cancer cells become resistant to drugs, for example.

To be able to mimic in vivo environments allows researchers to expand and culture next generation living organoid biobanks. And because they can be produced on a large scale, organoid biobanks provide more opportunities for toxicology studies and personalized and precision medicines.

HistoSpring Leads the Way in Tumor Organoids
HistoSpring is at the forefront of this advanced biotechnology and currently offers breast and ovarian organoids for advanced research.

The team cultivates and freezes breast and ovarian organoids and then sends them to researchers and engineers who further propagate for larger scale organoid growth. The researchers can perform a wide variety of testing on the organoids, such as ex-vivo testing of drug response.

The organoids offered by HistoSpring are invaluable to academic researchers and other research professionals who require tumor organoids that closely mimic the cancerous masses that actually grow inside patients and that respond to treatment in the same way as in vivo tumors do. Researchers who need the highest quality tumor organoids and highly personalized customer service turn to HistoSpring – and you can too.