Home Blog

Genome Doubling in Cancer Cells Involves Cell Division Glitch

Working with human breast and lung cells, Johns Hopkins Medicine scientists say they have charted a molecular pathway that can lure cells down a hazardous path of duplicating their genome too many times, a hallmark of cancer cells.

The findings reveal what goes wrong when a group of molecules and kinases are triggered to regulate the cell cycle, resulting in replication of chromosomes in cells that exit the cell cycle. The findings, the team suggest, could be used to develop therapies that interrupt these abnormal events in the cell cycle, and have the potential to stop the growth of cancers.

“An enduring question among scientists in the cancer field is: How do cancer cell genomes get so bad?” says Sergi Regot, PhD, associate professor of molecular biology and genetics at the Johns Hopkins University School of Medicine. “Our study challenges the fundamental knowledge of the cell cycle and makes us reevaluate our ideas about how the cycle is regulated.”

Regot and colleagues reported on their findings in Science, in a paper titled “CDK4/6 activity is required during G2 arrest to prevent stress-induced endoreplication.”

To replicate, cells follow an orderly routine that begins with making a copy of their entire genome. This is then followed by separation of the genome copies, and finally, the replicated DNA is divided evenly into two “daughter” cells. Human cells have 23 pairs of each chromosome—half from the mother and half from the father—including the X and Y sex chromosomes. This makes 46 total, but cancer cells are known to go through an intermediate state that has double that number—92 chromosomes. But how this happens was a mystery.

As the authors explained, abnormal DNA content is one of the most common characteristics of cancer cells, with recent evidence suggesting that 35 to 40 percent of tumors go through a whole-genome doubling (WGD) event as they evolve. “WGD is associated with genome instability, metastasis, and worse overall prognosis,” the team wrote, “However, the molecular mechanisms that drive WGD are poorly understood.”

Regot noted that cells that are stressed after copying the genome can enter a dormant, or senescent stage, and mistakenly run the risk of copying their genome again. And while generally and eventually, these dormant cells are swept away by the immune system after they are recognized as faulty, there are times, especially as humans age, when the immune system can’t clear the cells.

Left alone the abnormal cells can replicate their genome again, shuffle the chromosomes at the next division, and a growing cancer begins. A model known as endoreplication, the team explained in their paper, is when the cell undergoes two rounds of DNA replication without cell division. Endoreplication, they noted, contradicts the idea of irreversible commitment to cell division of cells entering the cell cycle. “We rationalized that although DNA replication and cell division are coupled under normal circumstances, there must be away to uncouple these events during times of cellular stress and break the commitment to cell division even after DNA replication has occurred.”

In an effort to pin down details of the molecular pathway that goes awry in the cell cycle, Regot and graduate research assistant Connor McKenney, who led the Johns Hopkins team, focused on human cells that line breast ducts and lung tissue. These cells generally divide at a more rapid pace than other cells in the body, increasing the opportunities to visualize the cell cycle. “

Regot’s lab specializes in imaging individual cells, making it especially suited to spot the very small percentage of cells that don’t enter the dormant stage and continue replicating their genome. For their newly reported study the team scrutinized thousands of images of single cells as they went through cell division. The researchers developed glowing biosensors to tag cyclin dependent kinases (CDKs), cellular enzymes that are known for their role in regulating the cell cycle. “Cell cycle events are coordinated by cyclin-dependent kinases (CDKs) to ensure robust cell division,” they wrote. “CDK4/6 and CDK2 regulate the growth 1 (G1) to synthesis (S) phase transition of the cell cycle …”.

Through their studies the team saw that a variety of CDKs activated at different times during the cell cycle. After the cells were exposed to an environmental stressor, such as a drug that disrupts protein production, UV radiation or osmotic stress (a sudden change in water pressure around cells), the researchers saw that CDK 4 and CDK 6 activity decreased.

Then, five to six hours later, when the cells started preparations to divide, CDK 2 was also inhibited. At that point, a protein complex called the anaphase promoting complex (APC) was activated during the phase just before mitosios—the step during which the cell pulls apart and divides. “In the stressed environment in the study, APC activation occurred before mitosis, when it’s usually been known to activate only during mitosis,” said Regot.

About 90 percent of breast and lung cells leave the cell cycle and enter a quiet state when exposed to any environmental stressors. In the scientists’ experimental cells not all of the cells went quiet. The research team discovered that five to ten percent of the breast and lung cells returned to the cell cycle, dividing their chromosomes again. “Upon relief of stress, a significant fraction of cells underwent a second round of DNA replication that led to whole-genome doubling,” they wrote.

Through another series of experiments the team linked an increase in activity of stress activated protein kinases (SAPKs) to the percentage of cells that skirt the quiet stage and continue to double their genome. “Mechanistically, G2 cell cycle exit occurs by premature activation of the anaphase-promoting complex or cyclosome (APC/C) in G2,” they wrote. “Notably, we found that premature APC/C reactivation occurs by persistent simultaneous inhibition of cyclin-dependent kinase 1 (CDK1) and CDK4/6 in G2 by SAPKs.”

The mechanisms, they noted, occurred independently of the p53 protein. “We found that SAPKs promote APC/C reactivation in G2 independently of p53.” This premature APC/C reactivation led to endoreplication of the genome in the small number of cells. “Stress-induced APC/C reactivation was driven by CDK4/6 inhibition during G2 arrest, challenging the classic definition of the restriction point and highlighting an important role of CDK4/6 beyond the G0 to G1 transition,” they noted.

Regot says there are ongoing clinical trials testing DNA-damaging agents with drugs that block CDK. “It’s possible that the combination of drugs may spur some cancer cells to duplicate their genome twice and generate the heterogeneity that ultimately confers drug resistance,” says Regot. “There may be drugs that can block APC from activating before mitosis to prevent cancer cells from replicating their genome twice and prevent tumor stage progression.” The authors further pointed out that SAPK signaling plays a central role in a variety of stress conditions that are related to aging, including proteotoxic stress, oxidative stress, and inflammation.

In the context of their study findings, they concluded, “Because SAPK-induced endoreplication is independent of p53 and most cancers with WGD lose p53 function prior to genome duplication, we propose that SAPK-mediated WGD might contribute to genetic instability and tumorigenesis in aging tissues.”

In an associated perspective in Science, Bart Westendorp, PhD, at Utrecht University, and Regenerative Medicine Center Utrecht, commented, “Given that CDK4/6 inhibition is now standard-of-care therapy for estrogen receptor–positive breast cancer, it is important to study whether these inhibitors may promote unwanted whole genome doubling events and genomic instability in patients … To answer such questions, innovative approaches will be needed for in vivo tracking of the long-term fates and clonal outgrowth of tetraploid cells arising from the G2 phases in which CDK4/6 activity is disrupted.”

Using Multiplexed Multi-Omics to Study Spatial Heterogeneity in Ovarian Cancer

Broadcast Date: 
  • Time: 

Studying the spatial organization of all the cells in the niche or neighborhood of a tumor is crucial to fully understand how cancers progress. Since proximity speaks to cellular activity, the spatial positioning of cells certainly plays a role in modulating clinical outcomes. However, most platforms used to reconstruct the tumor ecosystem (TE) fail to include spatial context in the three-dimensional (3D) space of a solid tumor with single-cell resolution, and thus lack information on cell-cell or cell-extracellular matrix interactions.

In this GEN webinar, Dr. Sammy Ferri-Borgogno will present the results of a study that provides insights into the molecular basis of spatial cell heterogeneity in ovarian cancer. During her presentation, she will present a pipeline of integrated multiplex multi-omics 3D spatially resolved modalities using FFPE gynecological tumor samples. The multi-omics modalities she will discuss include non-targeted mass spectrometry imaging, Stereo-seq, and targeted seqIF. You’ll learn how these spatially resolved modalities identify analytes in voxels across serial tissue sections, revealing an integrated 3D spatial map that displays cell identity, activation, and energized status.

A live Q&A session will follow the presentation, offering you a chance to pose questions to our expert panelist.

Sammy Ferri-Borgogno
Sammy Ferri-Borgogno, PhD
Instructor
Department of Gynecologic Oncology and Reproductive Medicine
MD Anderson Cancer Center

 

Endpoints: Critical Preclinical Markers in Neurodegeneration Disease Assays

Broadcast Date: 
  • Time: 

Altered dopaminergic neurotransmission underlies a variety of neurological and neuropsychiatric diseases, including playing a central role in the neurodegenerative processes that lead to Parkinson’s disease. Loss of dopaminergic neurons in the substantia nigra within the brain disrupts motor control pathways resulting in the development and progression of symptoms such as bradykinesia, rigidity, and tremors.

The degeneration of dopamine neurons is believed to result from neuronal dysregulation and cell death subsequent to oxidative stress and mitochondrial dysfunction related to lysosomal accumulation of a toxic form of alpha-synuclein. Understanding the complex interplay between alteration of dopamine signaling and neurodegeneration is crucial for developing targeted therapeutic strategies that preserve dopaminergic function to address Parkinsonian symptoms and ultimately identify appropriate neuronal pathways that may mitigate dopamine neuronal loss and disease progression.

In this GEN webinar, our speaker John Renger, PhD, chief scientific officer at Cerevel Therapeutics, discusses preclinical markers used in assays relevant to neurodegenerative disease processes, including the evaluation of autophagy and mitophagy using methods such as immunohistochemistry, western blotting, ELISA-like assays, high content analysis, and other platforms. You’ll gain insight into how these assays are able to yield important insights into disease progression models of neurodegeneration and reveal the potential of novel targets for new treatments to slow or stop progression of debilitating neurodegenerative disease.

A live Q&A session will follow the presentation, offering you a chance to pose questions to our expert panelist.

John Renger
John Renger, PhD
Chief Scientific Officer
Cerevel Therapeutics

 

BioSolution Designs and RoosterBio Collaborate on Engineered MSC-Derived Biotherapeutic Development

BioSolution Designs (BSD) reported that it is collaborating with RoosterBio to simplify the development and manufacturing of engineered human mesenchymal stem/stromal cell- (MSC) derived therapies.

The partnership leverages BSD’s multigenic design and assembly platform, Bird of Prey, and cell engineering expertise along with RoosterBio’s MSC and exosome bioprocessing products, manufacturing protocols, and analytical services, according to Thomas D. Reed, PhD, CEO of BSD, with the goal of delivering an end-to-end solution for the engineering and development of cell- and exosome-based therapeutics.

“Mesenchymal stem cells represent a major untapped resource with broad therapeutic utility, from regenerative medicine to treating rare diseases. Understanding all too well that ‘The Product is the Process,’ we are thrilled to collaborate with RoosterBio, whose raison d’etre revolves around controlling all aspects of scalable cGMP manufacturing and characterization of human MSCs and MSC-derived exosomes,” said Reed.

“This partnership will not only benefit our internal therapeutic initiatives but will provide a seamless transition for BSD’s OspreyBio customers to send multigenic therapeutic candidates to RoosterBio for efficient manufacturing.”

“By combining Biosolution Designs’ innovative multigenic technologies with our well-established cell therapy products and scalable bioprocess manufacturing platforms, we will accelerate the clinical progression of the next generation of cell and exosome therapeutics,” added Tim Kelly, CEO of RoosterBio.

Multispecific Antibodies Come to Grips with Manifold Targets

Antibodies perform like guided missiles, inspiring therapeutic developers to produce their own antibody-based weapons systems. Whereas natural antibodies have structures that are determined by the adaptive immune system, designed antibodies have structures that reflect human ingenuity. Ultimately, both natural and designed antibodies do the same thing: recognize pathogenic targets and help bring about their destruction.

Designed antibodies, however, have an advantage. Whereas natural antibodies are typically monospecific, designed antibodies may be multispecific; that is, they may be bi-, tri-, or even tetraspecific. In the target-rich environments presented by disease processes, multispecific antibodies may prove to be effective weapons systems, perhaps even more effective than current antibody-based therapeutics.

More than 100 antibody-based therapeutics have been approved for the treatment of diseases ranging from cancer to neurodegeneration. To build on this success, drug developers are advancing the development of multispecific antibodies. (Most of these new antibodies are bispecific, which means they are able to bind to two different targets at the same time.) Indeed, more than 100 bispecific antibody products are in clinical trials.

Several of the latest bispecific/multispecific antibody technologies are described in this article, which GEN prepared after speaking with several experts in the field of antibody design. For example, there are technologies for developing T-cell bispecific antibodies, which are novel cancer treatment candidates that can crosslink tumor cells and T cells. There are technologies to enhance immunoglobulin (IgG) hexamerization, the formation of six-antibody complexes that can be induced by cell surface antigens. There are technologies to concentrate radioactive therapy by using bispecific antibodies that simultaneously engage a tumor and a small radioactive peptide. Finally, there are technologies to move beyond bispecific antibodies and create multispecific antibodies that can better address the multifactorial traits of diseases.

Neoantigens as beacons

Despite the transforming impact of covalent small-molecule inhibitors, cancer drug resistance remains a daunting challenge. To take on this challenge, Aethon Therapeutics develops antibodies that can enhance the effects of small-molecule inhibitors, or targeted covalent inhibitors (TCIs), by alerting the immune system to TCI-generated neoantigens.

“Aethon unites cancer-specific covalent inhibitors with immunotherapy based on T-cell-engaging bispecific antibodies to make responses to targeted cancer therapies more effective, with a potential for cures,” says Raj Chopra, FRCP, FRCPath, FRSB, PhD, the company’s CEO. (He is also head of oncology and a venture partner at Apple Tree Partners, which has Aethon in its portfolio.)

Raj Chopra
Raj Chopra, FRCP, FRCPath, FRSB, PhD
CEO, Aethon Therapeutics

Aethon’s proprietary HapImmune platform was developed by its scientific co-founders at New York University (Hattori et al. Cancer Discov. 20239; 13: 132–145). “They found that covalent inhibition of target proteins caused by genetic mutations in oncogenes such as KRAS results in the cell surface presentation of a cancer-specific neoantigen drug-peptide on MHC molecules,” Chopra relates. “This MHC presentation of the drug-peptide conjugate is a beacon, if you will, that appears on the tumor cell but not on healthy cells. This beacon exists even when the cell is resistant to the original covalent inhibitor drug.”

Aethon creates specifically engineered bispecific antibodies targeting this drug-peptide MHC complex with high precision. “The beacon presented by the MHC helps the immune system detect and eradicate the persister and resistant cancer cells,” explains Christoph Rader, PhD, the company’s CTO. “It’s a one-two punch in which the first blow is targeted therapy and the second blow by the immune attack elicited by the Aethon drug.”

Christoph Rader
Christoph Rader, PhD
CTO, Aethon Therapeutics

The company’s proof-of-concept preclinical studies have focused on an oncogenic KRAS mutation frequently found in non-small cell lung cancer (NSCLC) and other cancers treated with the approved covalent inhibitors sotorasib and adagrasib. “To further enhance the covalent inhibitor’s activity, Aethon has developed companion T-cell-engaging bispecific antibodies targeting the drug-peptide MHC complex created by sotorasib and adagrasib,” Rader notes. “[The company] has confirmed their efficacy and selectivity in both in vitro and in vivo models of NSCLC.”

Chopra and Rader say because of its versatility, the platform will be applied to virtually any covalent drug that selectively modifies a cancer mutation and becomes targetable as an MHC-restricted synthetic neoantigen on the cancer surface.

Aethon HapImmune Platform illustration
When covalent inhibitors bind target proteins, they create drug-target peptide conjugates, or haptens, that can be presented on the surface of tumor cells. To create antibodies that can home in on haptens, Aethon Therapeutics applies its proprietary HapImmune platform. These antibodies are then reformatted into bispecific T-cell engagers that can selectively kill drug-treated cells.

Hexamer technology

Utilizing multiple antibody design approaches can expand therapeutic applications. “We have developed several technology platforms—including the DuoBody, HexaBody, DuoHexabody, and HexElect platforms—to generate novel antibody-based therapeutics that are designed to harness the power of human antibodies,” says Esther Breij, PhD, vice president, global head of translational research, Genmab. “[Our goal is to transform] how cancer and other serious diseases are treated.”

Esther Brei
Esther Breij, PhD
VP, Global Head of Translational Research, Genmab

The DuoBody platform generates bispecific antibodies suitable not only for discovery but also for commercial manufacturing scale using a sequential process. The process involves initial production of two IgG1 antibody molecules of the desired specificity that contain matched mutations in the constant domain (Fc domain). Following separate expression and purification, the two antibodies are recombined under a process called controlled Fab-arm exchange.

The HexaBody platform modifies monoclonal antibody to enhance target-mediated IgG hexamerization on the plasma membrane. “Combining these technologies can allow for the creation of even more potent therapeutics,” Breij suggests. “We are basically combining dual targeting with enhanced potency to increase antibody effector functions.”

The utility of the technology has been demonstrated in recent preclinical studies. For example, Genmab researchers and their colleagues used it to develop DuoHexaBody-CD37, a humanized bispecific IgG1 antibody that shows promise against B-cell malignancies (Oostindie et al. Blood Cancer J. 2020; 10: 30). This antibody binds two non-overlapping epitopes on CD37 with an engineered hexamerization-promoting mutation in the Fc region.

Genmab Duo Hexa Body
Genmab uses its DuoBody, HexaBody, DuoHexaBody, and HexElect technologies to build on natural antibody biology and transform how cancer and other serious diseases are treated. DuoBody molecules, bispecific molecules that bind to two non-overlapping epitopes, can be engineered with a hexamerization-promoting mutation in the Fc region. The result, a DuoHexaBody molecule, is shown here. It is a cluster of six bispecific antibodies with enhanced effector functions.

CD37 is a tetraspanin selectively expressed on B cells. Most monoclonal antibodies specific for CD37 are poor inducers of tumor killing by complement-dependent cytotoxicity. However, DuoHexaBody-CD37 showed efficient cytotoxicity-mediated tumor cell killing in primary tumor cells from patients with B-cell malignancies, and in leukemic cell lines and patient-derived xenograft models representing different B-cell malignancies.

Genmab has used the DuoBody platform as the basis of four approved bispecific antibody medicines. The company has about 20 products in clinical development that incorporate hexamer technology.

Safer radioimmunotherapy

Treating cancer with radioimmunotherapy involves the use of tumor-targeting antibodies that are labeled with radioisotopes. “However, the exposure of healthy tissue to radiation has been a major challenge,” advises Randolf Kerschbaumer, PhD, the CEO of OncoOne. “We have engineered our proprietary technology to address the challenges of antibodies directly linked to a cytotoxic payload, especially cytotoxic radionuclides, thereby minimizing the risk of healthy tissue exposure.”

Randolf Kerschbaumer
Randolf Kerschbaumer, PhD
CEO, OncoOne

“Our PreTarg-it platform is a two-component system for the pretargeted delivery of radionuclides to the tumor,” he continues. “The first component is a tailored bispecific antibody, in which one binding arm has a high specificity for a tumor target. The bispecific, nonradioactive antibody is applied first and accumulates at the tumor site expressing the target while unbound antibodies are cleared from the circulation.

“Once the bispecific antibody is cleared from circulation, a small peptide that is conjugated to a chelator loaded with a radioactive nuclide is applied. The peptide is then trapped in the tumor via the antibody’s second specificity, while the excess of unbound radioactive peptide is excreted within hours through the kidney and bladder. Thus, the cytotoxic activity is almost exclusively concentrated in the tumor.”

According to Kerschbaumer, the PreTarg-it platform has demonstrated its versatility and modularity by helping OncoOne generate four therapeutic candidates. “The bispecific antibody consists of modules that can be exchanged, thereby allowing us to generate bispecific antibodies capable of binding to various tumor targets,” he explains. “We can also conjugate peptides to different cytotoxic agents, like radionuclides, immunomodulators, and (encapsulated) toxins. Therefore, our platform enables us to target different tumor antigens with payloads of different substance classes.”

Currently, the PreTarg-it platform is being investigated in hard-to-treat cancers such as pancreatic cancer. “These diseases usually have a very poor prognosis and are often too far advanced when diagnosed to respond to conventional approaches such as immunotherapy,” Kerschbaumer observes. “We are advancing the platform currently through preclinical evaluation, where we have already demonstrated its ability to deliver strong efficacy within one treatment cycle.”

OncoOne PreTarg-it Technology
OncoOne has created the PreTarg-it platform, which employs a three-step radio-immunotherapy approach. First, one arm of a bispecific antibody targets a tumor marker. Second, the antibody is cleared from the bloodstream and concentrated within the tumor. Third, a radioligand is delivered that binds to the other arm of the antibody. Unbound radioligand is removed via the kidneys, reducing radiation exposure to healthy tissue.

Multispecifics to enhance potency

“Even though patients have a single disease, it may display multifactorial traits,” says Mark Chiu, PhD, president and CEO, Tavotek Biotherapeutics. “Thus, we engineer our molecules to have multiple mechanisms of action.” Tavotek accomplishes this task by using its multispecific antibody platform.

“While bispecific antibodies have two recognition arms, multispecific antibodies have more than two engaging domains,” Chiu points out. “The purpose of both antibody types is to accomplish multiple functions as mediated by interactions with disease targets.”

Chiu adds that a trispecific antibody may have two arms that bind cancer targets and a third arm with a unique function such as immune cell engagement to then kill the tumor cells. “Tavotek utilizes a repertoire of modules—including antibody-based binding arms, cytokines, and enzymes—to engineer multispecific antibodies with avidity that exploit dominant traits of multifactorial diseases to enhance therapeutic drug efficacy.”

According to Chiu, the design of bi-, tri-, tetra-, or multispecific molecules has the following steps: First, define a therapeutic mechanism of action. Second, generate the building blocks. (For this step, Tavotek has the TavoSelect platform.) Third, assemble the building blocks into a differentiated molecule with unique functions to generate the building blocks. (For this step, Tavotek has the TavoPrecise platform).

Tavotek is developing TAVO412, a multispecific antibody that is in a Phase I study for the treatment of advanced or metastatic solid tumors. TAVO412 targets mesenchymal-epithelial transition factor (cMET), epidermal growth factor receptor (EGFR), and vascular endothelial growth factor (VEGF).

Cancer often progresses because of the development of mutant forms of EGFR and enhanced crosstalk with cMET, remarks Maria MacWilliams, PhD, vice president of molecular biology at Tavotek. “TAVO412 pairs anti-EGFR and anti-cMET with anti-VEGF binding arms to enhance tumor resistance mitigation and shut down angiogenesis,” she adds. “This ‘trifecta’ approach extends TAVO412’s use to patients suffering from triple-negative breast cancer or from liver, gastric, or pancreatic cancer.”

Tavotek has oncology drug candidates in addition to TAVO412 in its pipeline. Also, the company is developing immunology drug candidates. Finally, as Chiu notes, the company has plans to develop multispecific antibodies that enable specific delivery of siRNA to diseased tissue. Possibilities include the control of master regulator gene expression, targeted engagement to activate or modulate immune cells, and intracellular targeting agents that can cross tissue barriers to facilitate various kinds of delivery (such as oral, pulmonary, and cerebral delivery).

ASGCT Preview: Mingozzi and Keeler Discuss the State of Gene Therapy

On last month’s episode of our monthly science talk show GEN Live“Is Gene Therapy Nearing a Tipping Point?”—I was joined by a pair of gene therapy experts to discuss the state of gene therapy, the promises and pitfalls of delivery vectors, the challenge posed by the immune response, and the safety issues that need to be addressed going forward. 

Allison Keeler, PhD, assistant professor at the University of Massachusetts Chan Medical School and the Gene Therapy Center at UMass, studies the immune responses to AAV gene therapy as well as developing novel immunotherapies by cell editing and engineering.  

Also joining the conversation was Federico Mingozzi, PhD, a veteran in the gene therapy field for more than two decades and the chief science and technology officer at Spark Therapeutics for the past six years. Before joining Spark, Mingozzi had a distinguished academic career, including stints at the Children’s Hospital of Philadelphia (CHOP), INSERM and Genethon in France. It was when he moved from Italy to the CHOP for a postdoc with Roland Herzog, PhD, (now at Indiana University School of Medicine) where his path crossed with gene therapy. He ended up working closely with Kathy High, MD, and got involved in the early studies of hemophilia gene therapy as well as conducting early work in ocular gene transfer in collaboration with Jean Bennett, MD, PhD, at the University of Pennsylvania—the work that was the basis for Luxturna. He is currently launching a new startup that is still in stealth mode. 

After we discussed AAV vectors, nonviral delivery methods, the immune response, the importance of biomanufacturing, and more, we turned the conversation to the upcoming American Society of Gene and Cell Therapy (ASGCT) meeting in Baltimore, where Mingozzi, Keeler, and myself are planning to spend the second week of May.  

Here’s what Keeler and Mingozzi are looking forward to at ASGCT 2024: 

Keeler: I am always interested in seeing the clinical trial updates, because we don’t always get to see that data. Going to ASGCT and seeing what is going on in the clinic always motivates everyone in the lab and reminds us why we’re doing the work that we do. It is really exciting, even if it is outside of our area. 

Both Federico and I sit on the immune responses panel, so I’m always excited to see the symposium that we put together, as well as the abstracts focusing on that topic. I’m interested to see some of the CRISPR talks. Those are usually really interesting talks. And there will probably be some machine learning talks as well. Those all fall under the things that will be new. But it’s also wonderful to see updates on clinical trials that we’ve seen year after year. That is usually my favorite part of the conference.

Mingozzi: I shouldn’t date myself but I have had the privilege to see the field over many years—where we were and where we are today: It’s incredible! Every year ASGCT is so exciting.  

gene therapy / ASGCT
Mingozzi, Keeler, and LeMieux on GEN Live

Like Allison, I’m looking forward to the clinical data. There are perhaps some newcomers—when it comes to AAV in the hearing space—where gene therapy can make a difference. There are some presentations around clinical trials for hearing disorders that I find exciting and an interesting parallel to where we were with the eye many years ago. And a testimony that the platform can still deliver important results in areas of high medical need. 

I’m also interested in the technology development that is brewing from non-viral delivery to the newest version of genetic editing and, of course, delivery, delivery, delivery! How we bring these new platforms to the tissue of interest. I think that’s still one of the big questions that genomic medicine is trying to get hold of it—sometimes successfully and sometimes still a work in progress.

Keeler: One thing I really enjoy at ASGCT is seeing all the trainees presenting. We try to give oral abstracts and short talks to a lot of the graduate students and postdocs working in the lab. And sometimes, I think those are the most exciting and interesting talks. For example, I have a graduate student that’s giving a talk on Saturday. It’s really great to see students and people from diverse backgrounds and all the trainees presenting their work and all the hard things that they’re doing in the lab. That is something that I really look forward to.

Mingozzi: I agree. That is the future of our work. And it’s fantastic to see how ASGCT is really reaching out to low- and medium-income countries. There is a lot of effort to bring gene therapy, both regarding education and also gene therapy per se, to those countries. It is fantastic to see the impact of what the society is doing worldwide.

LeMieux: Well now I’m even more excited to go than I was before! It’s going to be a great meeting. See you in Baltimore!   

You can watch the entire interview by registering here

Myelin Sheath Damage Mediated by Epigenetic Remedy in Multiple Sclerosis

Despite being one of the most common and well-known neurodegenerative diseases, multiple sclerosis (MS) remains difficult to treat. However, new research from scientists at Cincinnati Children’s Hospital Medical Center and elsewhere points to a potential therapeutic approach that appears to overcome difficulties faced by other attempts. Their work is described in a new Cell paper titled, “Small-molecule-induced epigenetic rejuvenation promotes SREBP condensation and overcomes barriers to CNS myelin regeneration.”

According to the paper, the researchers treated mouse models of multiple sclerosis and myelin organoids with an inhibitor molecule called epigenetic-silencing-inhibitor-1 (ESI1) that appears to improve both myelin production and poor cognitive function associated with MS and similar demyelinating diseases. It works at the epigenetic level to essentially restart myelin production by oligodendrocytes present in the MS lesions.

“These findings are significant as they offer new pathways for treatment that potentially shift the therapeutic focus from just managing symptoms to actively promoting repair and regeneration of myelin,” said Qing Richard Lu, PhD, scientific director of the Brain Tumor Center at Cincinnati Children’s and the study’s corresponding author. “Prior to finding ESI1, most scientists believed that remyelination failure in MS was due to the stalled development of precursors. Now we show a proof of concept that reversing the silencing activity in [oligodendrocytes] present in the damaged brain can enable myelin regeneration.” 

At the core of the findings is the observation that MS works by activating various cell types and signaling pathways that work together to silence the myelin repair process and prevent oligodendrocytes from doing their job. Analysis of stored autopsy tissues revealed that oligodendrocytes within MS lesions lacked an activating histone mark called H3K27ac, while expressing high levels of two other repressive histone marks H3K27me3 and H3K9me3 associated with silencing gene activity.

Armed with this information, the researchers searched small molecule libraries for compounds that could target enzymes responsible for modifying gene expression and influencing silenced oligodendrocytes. Of all the compounds considered, ESI1 proved nearly five times more powerful. In tests, it tripled the levels of the desired H3K27ac histone mark while sharply reducing levels of the two repressive histone marks. The compound also promotes the formation of biomolecular condensates in the nucleus that serve as regulatory hubs for controlling the essential fats and cholesterol needed to make myelin. 

When tested in both aging mice and mouse models of MS, treatment with ESI1 prompted myelin sheath production and improved lost neurological function. These results were based on tests tracking gene activation, measuring microscopic sheaths surrounding axons, and observing the behavior of treated mice. Similarly, when the organoid cultures were exposed to ESI1, the treatment extended the myelin sheath of myelinating cells. 

Myelin regeneration treatment could be a boon for people living with MS as well as people recovering from brain and spinal cord injuries. But the most far-reaching implication of the study is the possibility of using ESI1, or similar compounds, to help slow or even reverse cognitive losses that often occur during aging. 

However, more research is needed to determine whether human clinical trials can be launched to evaluate ESI1 as a potential treatment. For example, the effects of ESI1 may need to be modulated by adjusting the dose, treatment duration, or using “pulsed therapy” during specific time windows. More research is also needed to determine the feasibility of designing compounds even more effective than ESI1 from scratch. 

New Research Power Ups Psilocybin’s Potential as an Antidepressant

Depression affects an estimated 300 million people around the world and is the leading cause of disability. Therapeutics for depression are widely available. However, they have limited efficacy and can have serious adverse effects that are associated with low patient adherence. The naturally occurring serotonergic hallucinogen psilocybin found in several species of mushrooms has been discussed as a potential treatment for depression. Now, a team of researchers from the University of Oxford and the Northern Ireland Methodology Hub at Queen’s University sought to determine the efficacy of psilocybin as an antidepressant compared with placebo or non-psychoactive drugs. The researchers say their findings are encouraging but further research is needed to clarify the factors that maximize psilocybin’s treatment potential for symptoms of depression.

The findings are published in BMJ Today in an article titled, “Efficacy of psilocybin for treating symptoms of depression: systematic review and meta-analysis.”

The researchers examined databases looking for randomized controlled trials that compared psilocybin as a treatment for symptoms of depression with controls, such as placebo, niacin (vitamin B), or micro doses of psychedelics.

They found seven relevant trials for analysis involving 436 participants with depression (52% female; 90% white). Changes in depression scores were measured using a statistical method called Hedges’ g. A Hedges’ g of 0.2 indicates a small effect, 0.5 a moderate effect, and 0.8 or more a large effect.

The change in depression scores was significantly greater after treatment with psilocybin than with a comparator treatment, with an overall Hedge’s g of 1.64 indicating a large effect size favoring psilocybin.

Further analyses to account for trial differences indicated that having secondary depression (related to an underlying disease) rather than primary depression, being assessed with a self-reported scale rather than a clinician assessed scale, older age, and previous use of psychedelics, were correlated with greater improvements.

The study authors acknowledge that high levels of variation (heterogeneity) between trials resulted in a low certainty of evidence to support a strong antidepressant effect of psilocybin, and generalisability of findings were limited by the lack of participant diversity.

Furthermore, in clinical trials, patients receive psilocybin in a calm living room with soothing music, supervised by a psychotherapist, which is unlikely to be achievable in a healthcare system.

As such, the authors concluded that, although this review’s findings are encouraging for psilocybin’s potential as an effective antidepressant, issues such as cost, lack of regulatory guidelines, and legal safeguards associated with psilocybin treatment need to be dealt with before it can be established in clinical practice.

Furthermore, there is still ongoing debate on whether psychedelics can express antidepressant activity on their own rather than by assisting specific forms of psychotherapy.

The researchers concluded that these promising findings “support a prudent approach in both scholarly and public settings, because more and better evidence is needed before any clinical recommendation can be made about therapeutic use of psilocybin.”

For similar GEN stories related to psilocybin and psychedelics for depression treatment read “Psychedelics Coming into the Modern Age of Medicine,” “Clinical Study Shows Hallucinogenic Magic Mushroom Compound Psilocybin Relieves Depression,” and “No Hallucination: The Launch of Psychedelic Medicine.”

Vendor-Agnostic Automation for Cell and Gene Therapy Manufacturing

A novel approach to automating cell therapy manufacturing using industry-standard equipment can potentially reduce human errors, deliver higher quality product, and lower manufacturing costs by approximately 70 percent.

This robotic technique, developed by Multiply Labs, automates cell therapy manufacturing equipment, and can accommodate new equipment as instruments and processes evolve. Because Multiply Labs’ approach is vendor agnostic, manufacturers can simply add this layer of automation atop their existing processes. Therefore, there is no need to redesign the manufacturing process to fit the equipment, which sometimes occurs with proprietary automation systems.

“Our approach is not to design instruments, consumables, or reagents,” Fred Parietti, PhD, co-founder and CEO of Multiply Labs, tells GEN. “Our approach teaches robots to use the good manufacturing process (GMP)-proven instruments and consumable reagents that people already are using so their process stays substantially the same but automated.”

To do this, Multiply Labs partners with leaders in the biopharma equipment space, including GenScript, Cytiva, Thermo Fisher, Charles River Laboratories, Akron Bio, and Fedegari, among others.

Cell and gene therapy manufacturing, today, is largely artisanal. “It reminds me of mechanical, hand-crafted watches,” Parietti says. That’s why manufacturing a cell or gene therapy product costs a rough average of $100,000 to $300,000 per dose. When those costs are combined with the high costs of research and development, drug prices soar into the million-plus dollar range.

Labor accounts for about half the manufacturing costs, Parietti says. “Automating cell and gene therapy doesn’t eliminate jobs,” he points out. “Supervising scientists are still needed; they’re just not pipetting now.” Automation removes the mundane tasks and those which are most likely to contaminate processes.

How Robotics Improve Processes

Writing in Cytotherapy, Parietti, co-senior author Jonathan Esensten, MD, PhD, associate adjunct professor, University of California San Francisco (UCSF), and their team cultured CD8+ T cells, comparing standard, manual cell expansion manufacturing process to a process using the same instruments, but operated by robotic systems. Both the manual process and the automated one used the following GMP-proven instruments: the Thermo Scientific™ Heracell™ VIOS incubator, the Wilson Wolf G-Rex bioreactor, and the Cytiva Xuri bioreactor.

The scientists noted that although the quality of automated and manual versions was statistically comparable, the standard deviation was lower for the robotic process. “The range of variation was much narrower, (increasing) robustness and repeatability,” Parietti says. “We also saw a decrease in actual errors in the automated culture,” he adds. “The tests were performed by PhDs at UCSF, so this was not a matter of (a lack of) skill.”

robotic cluster
Robotic cluster seen from the outside. [Multiply Labs]
Automation has been able to replace tube welding, “which is very manual and very risky,” he says, with a robotic connector. Similar technology is applied to cell sampling, which is performed with a connector-based syringe without exposing the cells to the atmosphere. “Everything we do is in a closed system,” Parietti emphasizes.

Resuspension is another example. “We learned that every scientist has their own way of swirling the culture flask, and some obtain higher yields than others. When a pharma company visits, I tell them, ‘I need your highest-yield scientist, and I know you have one.’ Everyone laughs because everyone knows that person’s name,” he says. Multiply Labs records the motion that the scientist uses and replicates it with the robots. “From that point on, you’re guaranteed that only that trajectory will be executed by the robot.”

Notably, the Thermo Scientific™ VIOS Automated Access CO2 Incubator can support 18 products at once per incubator. In contrast, manual manufacturing typically supports one, and competitors’ systems support up to 16, he says. Automation also shrinks the necessary production space to 400 to 500 square feet, down from the 1,000 to 2000 square feet needed for human-operated workflows.

Transitioning to robotics

One of the challenges reported in the Cytotherapy paper was the need for customized cartridges. Because robots can’t easily manipulate soft and unstructured objects, Multiply Labs repackaged the consumable sets of bags and tubes into a rigid cartridge. This “allow(s) the robotic arm to connect tubes, transfer liquids in and out of bags, and move bioreactors that were originally designed for human hands,” the scientists wrote. Now, Parietti says, “All the robot needs to do is pick up the cartridge and put it on top of the instrument, and it clicks in.” Cartridges are available from the instrument’s manufacturer.

To transition to robotic cell and gene manufacturing, biomanufacturers first should “review their processes and ensure we have automated the instruments they’re using,” Parietti says. He estimates the company has automated 70-80 percent of the major processing instruments used for cell and gene manufacturing and says he expects to automate several more during the next few months.

“Our goal is not to automate every possible instrument,” Parietti says, but to concentrate on the leading instruments for GMP manufacturing. “We take a modular approach with our robots so you can add or swap out modules.”

Universal Flu Vaccine May No Longer Be a Long Shot

A schematic diagram of the influenza virus, showing the surface proteins hemagglutinin (blue, peanut shaped) and neuraminidase (red, flower shaped), to which antibodies attach during an immune response. A new vaccine from Duke helps the immune system target the stalk of the hemagglutinin protein, rather than its top.
A schematic diagram of the influenza virus, showing the surface proteins hemagglutinin (blue, peanut shaped) and neuraminidase (red, flower shaped), to which antibodies attach during an immune response. A new vaccine from Duke helps the immune system target the stalk of the hemagglutinin protein, rather than its top. [U.S. Centers for Disease Control]

The search for a universal flu vaccine has been long and fraught with false starts and dead ends. For more than five years, Nicholas Heaton, PhD, associate professor of molecular genetics and microbiology at Duke University, has been working with his team on a systematic approach to create such a vaccine. One particularly promising approach is to develop antibodies that target both the head and stalk of the most abundant surface protein on the influenza virus. 

The group’s new study, entitled, “Vaccination with Antigenically Complex Hemagglutinin Mixtures Confers Broad Protection From Influenza Disease,” was published in Science Translational Medicine.  

Flu is widespread and deadly, with anywhere from 9–41 million cases annually, resulting in approximately 5–51,000 thousand deaths a year in the United States, according to the Centers for Disease Control and Prevention (CDC). The severity of flu infections is as variable as the strains that cause the disease. Each strain is typically characterized and referred to by alphanumeric codes including the letters H (hemagglutinin) and N (neuraminidase), identifying the most prominent surface proteins, and numbers to delimit the protein variant. The influenza virus contains about 5–10 times more hemagglutinin than neuraminidase, said Heaton. 

Current annual vaccines typically target the globular head of the lollipop-shaped hemagglutinin glycoprotein or neuraminidase proteins coating the virus. These proteins are highly mutable, resulting in the need to create new vaccines annually.  

“The virus has evolved to have the immune system recognize these (features on the head region). But these are the shapes the virus can change. That is an insidious strategy,” Heaton said. Furthermore, due to the sheer quantity of flu strains, only some of them are included in each year’s flu shot.  

For decades, researchers have sought out strategies to circumvent the rapid incessant evolution of flu strains. These strategies have varied from focusing on neuraminidase, to using stalk-targeting nanoparticles, or using live, genetically altered viruses.  

To broaden the function of flu vaccines, Heaton’s team now reports the development of “an antigenically complex mixture of recombinant hemagglutinins designed to redirect immune responses to more conserved domains of the protein.” Asked how this approach is different from previous efforts, Heaton told GEN, “I think that all (including ours) have their pros and cons.” 

As there are more hemagglutinin than neuraminidase molecules on the flu virus surface, the team focused their efforts on identifying regions of that protein that were less likely to mutate over time. “A number of groups have gone through and experimentally mutagenized the whole hemagglutinin and asked, ‘Which areas can change and still allow the hemagglutinin to function?’” Heaton explained. “The answer is, you can’t really change the stalk and expect it to continue to function.” 

“Antibodies against the stalk work differently,” Heaton explained. “Their mechanism of protection is not necessarily to block the first step of infection. So our idea was: What if we can come up with a vaccine that gives us both? What if we can get good head antibodies and at the same time also get stalk antibodies in case the vaccine selection was wrong, or if there’s a pandemic?” 

First, Heaton’s team aimed to create a group of mutated hemagglutinin proteins with a wide variety of mutations only in the head region, but with conserved stalks. They used gene editing techniques to create more than 80,000 variants of hemagglutinin with changes just in the head region after purification and sequencing procedures. They next verified that the mutated region was not prone to antibody recognition compared to the wildtype, suggesting that they had successfully generated a mutant protein mixture that could be used for vaccine creation.  

They then tested their mixture in animal models—mice and ferrets—to determine the antibody response. “If we took your blood to see if you are likely to be protected from a strain of flu, we’d be measuring what your antibodies do to hemagglutinin as the best metric of what’s likely to happen to you. The strongest correlates of protection have to do with hemagglutinin-directed immunity,” Heaton explained.   

Following this premise, the team found that animals vaccinated with the experimental mixture produced more antibodies against conserved stalk regions compared with controls. “One thing that we observed was enhanced vaccine-induced responses to both the head and stalk domain of the hemagglutinin,” Heaton told GEN. “That has been a difficult goal to achieve.” 

There are many avenues of further study ahead. “One of the most important next steps will be to understand if we can apply this approach to all of the different subtypes of influenza that infect humans. In this paper, we only study H1N1 viruses,” Heaton told GEN. 

Translating this data to humans will be a much larger hurdle. Heaton concluded, “There are many challenges in the advanced development of a vaccine. One major challenge is predicting how different people (with different immune exposure histories) will respond to the vaccine. That is very hard to model in a laboratory environment.”

chromosomes
Scientists charted a molecular pathway that can lure cells to duplicate their genome too many times, a hallmark of cancer cells. Their in vitro findings provide insights into mechanisms involving kinase enzymes that result in replication of chromosomes in cells that exit the cell cycle. The findings, the team suggest, could be used to develop therapies that interrupt these abnormal events in the cell cycle, and have the potential to stop the growth of cancers.
Neuromuscular Junction
In this GEN webinar, our speaker John Renger, PhD, CSO at Cerevel Therapeutics, discusses preclinical markers used in assays relevant to neurodegenerative disease processes, including the evaluation of autophagy and mitophagy using methods such as immunohistochemistry, western blotting, ELISA-like assays, high content analysis, and other platforms.
Mesenchymal stem cells
The partnership was established to leverage BioSolution Designs’ (BSD) multigenic design and assembly platform, Bird of Prey, and cell engineering expertise along with RoosterBio’s mesenchymal stem cell and exosome bioprocessing products, manufacturing protocols, and analytical services, according to Thomas D. Reed, PhD, CEO of BSD, with the goal of delivering an end-to-end solution for the engineering and development of cell- and exosome-based therapeutics.
Bispecific antibody in action
To build on the success of monospecific antibodies, drug developers are exploring the possibilities offered by bi-, tri-, and even tetraspecific antibodies.
Adeno-associated virus, illustration - stock photo
On a recent episode of GEN Live, Federico Mingozzi, PhD, joined Allison Keeler, PhD, to discuss the state of gene therapy. They shared their perspectives on the promises (and pitfalls) of delivery vectors, the challenges posed by the immune response, and innovations being made in both areas. They will both be at ASGCT next week, and shared what they are looking forward to at the meeting.
An image showing a myelin producing after treatment with ESI1 in mice
New research from scientists at Cincinnati Children’s Hospital Medical Center and elsewhere indicates that an inhibitor compound called epigenetic-silencing-inhibitor-1 or ESI1 could potentially improve myelin production and poor cognitive function associated with multiple sclerosis and other condition associated with demyelinating.
Magic Mushrooms
A team of researchers from the University of Oxford and the Northern Ireland Methodology Hub at Queen's University sought to determine the efficacy of psilocybin as an antidepressant compared with placebo or non-psychoactive drugs. The researchers say their findings are encouraging but further research is needed to clarify the factors that maximize psilocybin's treatment potential for symptoms of depression.
Regenerative Medicine
The robotic approach developed by Multiply Labs automates cell therapy manufacturing equipment and operations and can accommodate new equipment as instruments and processes evolve. Because Multiply Labs’ approach is vendor agnostic, manufacturers can simply add this layer of automation atop their existing processes. Therefore, there is no need to redesign the manufacturing process to fit the equipment, which sometimes occurs with proprietary automation systems
Influenza virus
The search for a universal flu vaccine has been long and fraught with false starts and dead ends. One particular promising approach is to develop antibodies that target both the head and stalk of the most abundant surface protein on the influenza virus.