Category Archives: Somatic Stem Cells

Outlook on the Worldwide Hunter Syndrome Industry to 2030 – ResearchAndMarkets.com – Yahoo Finance

The "Hunter Syndrome - Market Insights, Epidemiology and Market Forecast - 2030" drug pipelines has been added to ResearchAndMarkets.com's offering.

This report delivers an in-depth understanding of the Hunter Syndrome, historical and forecasted epidemiology as well as the Hunter Syndrome market trends in the United States, EU5 (Germany, Spain, Italy, France, and United Kingdom) and Japan.

The Hunter Syndrome market report provides current treatment practices, emerging drugs, and market share of the individual therapies, current and forecasted 7MM Hunter Syndrome market size from 2017 to 2030. The report also covers current Hunter Syndrome treatment practice/algorithm, market drivers, market barriers and unmet medical needs to curate the best of the opportunities and assesses the underlying potential of the market.

Hunter Syndrome Diagnosis

The diagnosis of Hunter syndrome is established in a male by identifying the deficient iduronate 2-sulfatase (I2S) enzyme activity in white cells, fibroblasts, or plasma in the presence of normal activity of at least one other sulfatase. Detection of a hemizygous pathogenic variant in IDS confirms the diagnosis in a male with an unusual phenotype or a phenotype that does not match the results of GAG testing. The diagnosis of this indication is usually established in a female with suggestive clinical features by identification of a heterozygous IDS pathogenic variant on molecular genetic testing.

Although the disease is almost exclusively reported in males, rare cases in females also do occur. The diagnosis of MPS II is usually established in a female patient with suggestive clinical features, such as the identification of a heterozygous IDS pathogenic variant on molecular genetic testing.

Molecular genetic testing approaches can include a combination of gene-targeted testing (single-gene testing, multigene panel) and comprehensive genomic testing (exome sequencing, genome sequencing) depending on the phenotype. Gene-targeted testing requires that the clinician determine which gene(s) are likely involved, whereas genomic testing does not.

Hunter Syndrome Treatment

Even with the introduction of ERT, patients with MPS II still require supportive symptomatic treatment from a wide range of specialists. A comprehensive initial assessment of each patient at diagnosis should, therefore, be undertaken, and should be followed by regular reviews. Supportive management and the anticipation of possible complications can greatly improve the quality of life of affected individuals and their families. Family members should be offered genetic counselling, and contact with other affected families, patients, and support groups.

It is now a decade since ERT with intravenous idursulfase (Elaprase), a recombinant form of human iduronate 2-sulfatase, has been approved in the United States and the European Union at a weekly dose of 0.5 mg/kg for the treatment of MPS II. The approval was mainly based on the results from a first trial on individuals with the slowly progressive form of the disease. In the following year several other studies were undertaken to investigate clinical safety and efficacy of ERT; these clearly showed that idursulfase has positive effects on functional capacity (distance walked in six minutes and forced vital capacity), liver and spleen volumes, and urine GAGs excretion. Recently, a 3.5-year independent study determined that long-term use of ERT is similarly effective in young (age 1.6-12 years at the start of ERT) and older individuals (age 12-27 years at the start of ERT). In addition, two recent studies have confirmed ERT efficacy in improving somatic signs and symptoms of the disease in all individuals, including infants younger than age 1 year and individuals with the early progressive MPS II phenotype.

Pretreatment with anti-inflammatory drugs or antihistamines, as is often done for ERT in other conditions, is not suggested on the label for Elaprase; however, if mild or moderate infusion reactions (e.g., dyspnea, urticaria, or systolic blood pressure changes of 20 mm Hg) cannot be ameliorated by slowing the infusion rate, the addition of treatment one hour before infusion with diphenhydramine and acetaminophen (or ibuprofen) to the regimen usually resolves the problem. Pretreatment can typically be discontinued after 6-10 weeks.

Story continues

Hematopoietic stem cell transplantation (HSCT) using umbilical cord blood or bone marrow is a potential way of providing sufficient enzyme activity to slow or stop the progression of the disease, however, the use of HSCT is controversial because of the associated high risk of morbidity and mortality. The use of HSCT has been controversial because of limited information regarding the long-term outcomes and the associated high risk of morbidity and mortality.

Scope of the Report

Reasons to Buy

For more information about this drug pipelines report visit https://www.researchandmarkets.com/r/8glsqs

View source version on businesswire.com: https://www.businesswire.com/news/home/20200807005485/en/

Contacts

ResearchAndMarkets.com Laura Wood, Senior Press Manager press@researchandmarkets.com

For E.S.T Office Hours Call 1-917-300-0470 For U.S./CAN Toll Free Call 1-800-526-8630 For GMT Office Hours Call +353-1-416-8900

Read the original here:
Outlook on the Worldwide Hunter Syndrome Industry to 2030 - ResearchAndMarkets.com - Yahoo Finance

Edited Transcript of CLDX.OQ earnings conference call or presentation 6-Aug-20 8:30pm GMT – Yahoo Finance

NEEDHAM Aug 7, 2020 (Thomson StreetEvents) -- Edited Transcript of Celldex Therapeutics Inc earnings conference call or presentation Thursday, August 6, 2020 at 8:30:00pm GMT

* Anthony S. Marucci

Celldex Therapeutics, Inc. - Founder, President, CEO & Director

Celldex Therapeutics, Inc. - SVP of Regulatory Affairs

Celldex Therapeutics, Inc. - Senior VP, CFO, Secretary & Treasurer

Celldex Therapeutics, Inc. - SVP of Corporate Affairs & Administration

Welcome to the Celldex Therapeutics Mid-Year 2020 Conference Call. My name is James and I'll be your operator for today's call. (Operator Instructions).

And then I'd like to turn the call over to Sarah Cavanaugh. Sarah, you may begin.

Sarah Cavanaugh, Celldex Therapeutics, Inc. - SVP of Corporate Affairs & Administration [2]

Thank you very much. Good afternoon and thank you all for joining us.

With me on the call today are Anthony Marucci, co-founder, President and CEO of Celldex, Dr. Tibor Keler, co-founder Executive Vice President and Chief Scientific Officer, Dr Diane Young, Senior Vice President and Chief Medical Officer. Sam Martin, Senior Vice President and Chief Financial Officer, Dr Margo Heath-Chiozzi, Senior Vice President of Regulatory and Dr Diego Alvarado, Senior Director of Research.

Before we begin our discussion, I'd like to mention that today's speakers will be making forward-looking statements. Such statements reflect on current views with respect to future events and are based on assumptions and subject to risks and uncertainties that could cause actual results to differ materially from those expressed or implied by such forward-looking statements. Certain of the factors that might cause Celldex's actual results to differ materially from those in the forward-looking statements, include those set forth under the headings Risk Factors and Management's Discussion and Analysis of Financial Condition and Results of Operation in Celldex's annual report on Form 10-K, quarterly reports on Form 10-Q and its current reports on Form 8-K as well as those described in Celldex's other filings with the SEC and its press releases.

All forward-looking statements are expressly qualified in their entirety by this cautionary notice. You should carefully review all of these factors and be aware that there may be other factors that could cause these differences. These forward-looking statements are based on information, plans and estimates as of this call, and Celldex does not promise to update any forward-looking statements to reflect changes in underlying assumptions or factors, new information, future events or other changes.

Please be advised that the question and answer period will be held at the close of the call. I'd also like to mention that because of the current COVID-19 situation and also two of our offices are located in the areas of the hurricane, we do have folks dialing in from a number of different remote locations and I ask that you may be bear with us phone lines are a little scratchy because we're dealing with multiple issues on that end.

So with that, I'd like to turn the call over to Anthony. Anthony?

Anthony S. Marucci, Celldex Therapeutics, Inc. - Founder, President, CEO & Director [3]

--------------------------------------------------------------------------------

Thank you, Sarah. Good afternoon everyone and thank you for joining us. We hope you are all safe and healthy and appreciate you're taking the time to connect with us today. We are looking forward to updating all of you on our progress and providing more detail on our plans for the future. I want to take a few minutes to review the recent events and then I will ask Diane to update you on our clinical programs and Sam to review the financials. We will close the call with your questions.

As you may likely know in early June of this year, Dr Marcus Maurer a leading medical expert in Urticaria, whose research focuses on mast cells presented data from our KIT inhibitor, CDX-0159 and a late breaking session at the EAACI Annual Congress. These data provided an important proof of concept for the program and suggested significant potential which will dramatically impact mast cell driven disorders. These data also help support the $150 million public offering driven by high quality healthcare investors. Importantly, these proceeds will fund the company through 2023 and a number of very important milestones. We are on track to initiate two studies of CDX-0159 and chronic urticaria this fall and I have completed considerable work that Diane will discuss the support expanded development in 2021 and beyond.

As we have always done, we believe is important to focus our resources of people and financial on the programs that hold the most promise for patients and shareholders. Based on the current data we have in-house, we have prioritized the development of our KIT inhibitor CDX-0159, our CDX agonist, CDX-1140 and the first candidate from our bispecific program CDX-527 which combines our proprietary CD-27 agonist with the PD-1 blockade. In turn, we have made a decision not to advance our ErbB3 inhibitor, CDX-3379, which has been in an exploratory study with cetuximab to assess the utility of biomarkers for patient selection and cetuximab resistant head and neck cancer. Despite prophylactic treatment which Diane will discuss in more detail, patients continue to have difficulty tolerating therapy and we believe our resources are best utilized to expand the development of CDX- 0159 and our other pipeline programs.

For our CDX-0159 program, we intend to start two urticaria studies, one in inducible urticaria and the other in spontaneous urticaria this fall and to initiate both the Phase 1 study of CDX- 527 and refractory advanced cancers as well as the combination cohort of CDX-1140 with chemotherapy and treatment of naive metastatic pancreatic cancer later this year. This program is all support multiple data readouts later this year and next year including results from the CDX-0159 study and inducible urticaria in the first quarter of 2021 and the results from the study in spontaneous urticaria in the second half of next year.

We are also in the midst of a thorough assessment of additional opportunities for CDX-0159 and as we now of this list, we plan to initiate our third study and another mast cell driven disease next summer.

With this introduction, I would like Diane to cover activities in more detail. Diane?

--------------------------------------------------------------------------------

Diane C. Young, [4]

--------------------------------------------------------------------------------

Thank you, Anthony. Let me start with CDX-0159. CDX-0159 is a humanized monoclonal antibody developed by Celldex that binds to the KIT receptor with high specificity and potently inhibits it's activity. The KIT receptor tyrosine kinase is expressed in mast cells which mediate inflammatory responses such as hypersensitivity an allergic reaction. Ultimately, KIT signaling controls the differentiation, tissue recoupment, survival and activity of mast cells and we believe targeting KIT represents a unique strategy in diseases involving mast cells.

At the EAACI meetings, results from our recently completed Phase 1A study in healthy volunteers were presented. CDX -0159 demonstrated a favorable safety profile as well as profound and durable reductions of plasma tryptase, a protease made almost exclusively by mast cells. The phase 1A study was a randomized double blind, placebo-controlled single-ascending dose escalation study of CDX-0159 in 32 healthy subjects.

Subjects received a single intravenous infusion of CDX-0159 at 0.31319 milligrams per kilogram or placebo. As Dr. Maurer presented a single dose of CDX-0159 supress plasma tryptase levels in a dose-dependent manner indicative of systemic mast cell suppression or ablation, tryptase reduction was evident at 24 hours after infusions and minimal levels were typically observed within one week. Tryptase suppression below the level of detection was observed after a single one milligram per kilogram dose and was maintained for more than two months at single doses of both 3 and 9 milligrams per kilogram.

A subset of subjects from the 3 milligram per kilogram and 9 milligrams per kilogram cohorts agreed to continued follow-up For tryptase analysis which was ongoing at the time of the EAACI meeting. This follow-up and analysis was completed in July and tryptase levels remain below the level of detection for 14 weeks in the 3 milligram per kilogram cohorts for 50% of the returning subjects and 18 weeks in the 9 milligrams per kilogram cohort for all returning subjects. In this study, dose-dependent increases in plasma stem cell factor also mere decreases in tryptase consistent with allosteric blockade of stem cell factor to KIT and further demonstrating complete target engagement in vivo.

Importantly, CDX-0159 also demonstrated a favorable safety profile. The most common adverse events were mild infusion related reactions which spontaneously resolved without intervention. Asymptomatic decreases in neutrophil and white blood cell counts were also observed in laboratory testings but we returning towards normal at the end of the study. We also observed long serum half-life and lack of anti-drug antibodies which provide support to explore less frequent dosing in future studies. Based on these results, we plan to initiate 2 Phase 1B studies of CDX-0159 this fall, one in chronic inducible urticaria and one in chronic spontaneous urticaria, both of which are mast cell driven diseases specifically selected to provide clinical proof of concept for CDX-0159. I'll start with the study in the inducible urticaria as this indication will read out first with data expected in the first quarter of next year.

There were multiple forms of inducible urticaria and 0.5% of the total population suffer from them. We have selected two of the most common forms: symptomatic dermagraftism and cold-induced urticaria. Symptomatic dermagraftism is characterized by the development of a wheel and flare reaction in response to a stroking, scratching or rubbing of the skin usually occurring within minutes of the inciting stimulus. People afflicted with cold-induced urticaria experienced symptoms like itching, burning wheels and angioedema where their skin comes in contact with temperatures below skin temperature. For both of these diseases mast cell activation, leading to release of soluble mediators is thought to be the driving mechanism leading to the wheels and other symptom.

As you can tell based on their name, what's unique about these indications Is that they are induced by certain triggers and importantly investigators can induce these same reactions in the clinic. Dr Maurer will lead this study in his specialty clinic for urticaria in Berlin. We expect to enroll 20 patients, ten with symptomatic dermagraftism and ten with cold-induced urticaria who are resistant to antihistamine treatment. Their symptoms will be introduced in the clinic and a single dose of CDX-0159 at 3 milligram per kilogram will be administered. Patients will be followed for 12 weeks to evaluate safety and tolerability, clinical activity and pharmacokinetics and pharmacodynamics. Importantly, we intend to perform serial skin biopsies on patients so we can explore the impact of CDX-0159 on mast cells in the skin. This will help address whether CDX-0159 is inactivating the mast cells or leading to their deaths in elimination from skin.

The second study will be in chronic spontaneous urticaria or CSU, an indication where patients experience urticaria symptoms without identification of a known cause. This is a disease driven by mast cell activation, the release of mediators resulting episodes of itchy hives, swelling and inflammation of the skin that can go on for years or even decades. It is one of the most frequent dermatologic diseases with the prevalence of 0.5% to 1% of the total population and up to 3.2 million cases annually in the US. The study will be a randomized, double-blind, placebo-controlled Phase 1B dose escalation study that includes patients who are still symptomatic despite antihistamine therapy. We expect to enroll 40 patients across four cohorts, who will receive CDX-0159 or placebo. The dose and dosing schedule will vary by cohort.

Patients dosed at 0.5 and 1.5 milligram per kilogram will receive three doses at 4-week intervals and patients dosed at 3 and 4.5 milligrams per kilogram will receive two doses at an 8-week interval. The 12-week treatment period will be followed by another 12 weeks of follow-up. So 24 weeks total. This design will provide necessary data on the safety of multiple doses and also allow us to evaluate the potential clinical activity of CDX-0159 in this patient population. Again, we will be evaluating safety and tolerability, symptomatic relief as measured through disease activity scores and pharmacokinetics and pharmacodynamics. The study will be conducted at 4 to 5 centers in the USA, beginning in the fall of 2020. We anticipate results from this study in the second half of next year.

For both inducible and spontaneous urticaria, it is clear that these patients can truly suffer. The two top complaints are constant intense itch and poor self image. Their symptoms prevent regular sleep, interfere with daily life and work activities which subsequently promote social withdrawal, isolation and depression. There is truly an unmet need for efficacious therapies that address the root cause of their disease, mast cells.

Beyond urticaria, there are many diseases in which mast cells are the principal driver or a thought to significantly contribute to the pathology. We are digging deeply into the potential opportunity for CDX-0159 in these indications to select additional areas for expansion. Our evaluation includes review of scientific literature, medical guidelines, regulatory documents and market analyzes and discussions with medical experts. We are prioritizing indications in which there is strong evidence that mast cells play an important role in pathophysiology where there are unmet medical needs and where we can envision a clinical development path with clear early decision point.

We have narrowed what began as a list of over 50 indications to 4 major areas of focus. Mast cell activation syndromes including mastocytosis, Asthma including severe forms of asthma, allergic asthma and exercise induced asthma, allergic conditions including food allergies and allergy mediated dermatologic conditions and mast cell driven gastrointestinal disorders.

Our next step is to lay out the clinical development and regulatory path as well as commercial opportunities to help in the final indication selection. We will also be monitoring the field closely to ensure our plans continually reflect all available scientific clinical regulatory and competitive data. Certainly as data begin to emerge from the urticaria studies, this will also inform our final decision. We will continue to update you as we complete our diligent but are confident we will be in a position to initiate a Phase 1B-2 study in a third indication by summer 2021.

Finally, in closing for CDX-0159 I want to point out that we have initiated formulation work for subcutaneous delivery which we believe will be important to the candidates future success. We believe we are well positioned given CDX-0159 and enhanced PK profile and the durable tryptase suppression we observed even at low doses. The preliminary feasibility studies at 150 milligrams per mill look promising.

With that overview on CDX-0159, let me turn now to CDX 1140 and CDX-527. CDX-1140 is a Celldex developed human agonist anti-CD-40 monoclonal antibody that was specifically designed to balance good systemic exposure and safety with potent biological activity a profile, which differentiates CDX-1140 from other CD-40 activating antibodies for systemic therapy. CD-40 expressed on dendritic cells and other antigen presenting cells is an important target for Immunotherapy as it plays a critical role in the activation of innate and adaptive immune responses.

CDX-1140 completed dose escalation as monotherapy and in combination with CDX-301 a dendritic cell growth factor in an ongoing Phase 1 study in patients with recurrent, locally advanced or metastatic solid tumors and B-cell lymphomas. A critical goal of this study was to achieve dosing levels that provide good systemic exposure without dose limiting toxicity. As reported at the SITC meeting last November, CDX-1140 reach this goal with the maximum tolerated dose and recommended Phase 2 dose of 1.5 milligrams per kilogram, one of the highest systemic dose levels in the CD-40 agonist class.

We believe the relatively low doses of other potent CD-40 agonist antibodies tested in the clinic to date may limit their potential and modifying in the tumor micro environment and are hopeful that CDX-1140 at this dose level will better penetrate tumor and be more impactful. Importantly, from a safety perspective at 1.5 milligram per kilogram CDX-1140 is associated with manageable immune related adverse events that are consistent with those observed with approved effective therapies like checkpoint inhibitors.

While CDX-1140 has shown promising signs of single agent activity, it's clear that the combination approaches that target multiple pathways in the immune system likely offer patients the best opportunities for improvement. To that end, we have added multiple combination expansion cohorts including with KEYTRUDA in patients who have progressed on checkpoint therapy and with CDX-301 in patients with head and neck squamous cell carcinoma.

We also expect to initiate a combination with standard of care chemotherapy in first-line metastatic pancreatic cancer later this year. An indication, we are very interested in because both preclinical and clinical data suggests that the CD-40 pathway may have important anti-tumor potential in this disease.

We also expect to report on interim data from CDX-1140 this fall, that would focus on data from the monotherapy expansion cohorts in squamous cell head and neck cancer and renal cell carcinoma, data from the combination with CDX-301 and preliminary data from the combination with KEYTRUDA.

CDX-527, our first bispecific antibody program is also expected to enter the clinic later this year. CDX-527 combines CD-27 mediated T-cell activation with PD-1 blockade. We have developed CDX-527 from our proprietary highly active PDL-1 and CD-27 human antibodies and demonstrated the bispecific to be more potent than the combination of the individual antibodies in preclinical models.

Importantly, our prior clinical experience combining the CD27 agonist antibody varlilumab with PD-1 blockade supports the integration of these two antibodies from a dosing safety and activity perspective. We would expect initial data from this program in the first half of 2021.

Before I turn the call over to Sam to discuss the financials, I want to provide a little more clinical context surrounding the decision on CDX-3379 development.

At ASCO 2019, we presented a retrospective analysis that suggested that the anti-tumor activity with CDX-3379 might be associated with somatic mutations in particular genes associated with tumor suppression.

We decided to examine this hypothesis in an exploratory manner in the ongoing trial to see if there was a path forward that would allow us to utilize biomarkers to identify a targeted population that would respond to CDX-3379. In parallel, we knew that we needed to improve the tolerability of the combination of CDX-3379 and cetuximab specifically diarrhea management. Unfortunately, despite diarrhea prophylaxis measures, this continue to be a side effect which in addition to severe skin rash caused dose reductions and delays in the majority of patients, making it difficult to achieve clinical benefit. When considered together and after talking to our study investigators, we believe the risk benefit profile does not support further development in patients and that the resources allocated to this program would be best focused on expanded development of CDX-0159 CDX-1140 and CDX-527.

We will also continue to advance our preclinical pipeline which is exploring several interesting targets including AXL IoT-4, CD 24 and cyclic-15. Updates on our preclinical programs will be presented at scientific meetings later this year and next.

In summary, we are very pleased with the progress we've made so far this year. We believe CDX-0159 has the potential to be a field changing product across multiple mast cell driven indications and that CDX-1140 is establishing itself as a clearly differentiated CD-40 agonist. We're excited to bring CDX 527 into the clinic and all combined, look forward to a very busy rest of 2020.

We continue to be mindful of COVID-19 and our partnering closely with our clinical trial sites to mitigate any COVID related impact on our studies. So far. we have been very successful in these efforts but like everyone else we are looking cautiously at this fall and winter and contingency planning to help mitigate any risk to our timeline.

With that, I thank you for your time and I will hand the call over to Sam to review the financials. Sam?

--------------------------------------------------------------------------------

Sam Martin, Celldex Therapeutics, Inc. - Senior VP, CFO, Secretary & Treasurer [5]

--------------------------------------------------------------------------------

Thank you, Diane. For the second quarter of 2020 net loss was $11 million or $0.50 per share compared to a net loss of $11.8 million or $0.84 per share for the second quarter of 2019. Net loss for the six months ended June 30, 2020 was $23.7 million or $1.20 per share compared to 29 million or $2.21 per share for the comparable period in 2019.

Research and development expenses were $21.4 million for the six months ended June 30, 2020 compared to $21.2 million for the comparable period in 2019.

General and administrative expenses were $7.2 million for the six months ended June 30, 2020 compared to $8.8 million for the comparable period in 2019.

As of June 30, 2020, we reported cash, cash equivalents and marketable securities of $206.9 million compared to $53.7 million as of March 31, 2020. The increase was primarily driven by net proceeds of $141.4 million from our June 2020 underwritten public offering and net proceeds of $23.7 million from sales of common stock under our controlled equity offering agreement with Cantor completed in the second quarter prior to the public offering in June.

These increases were offset by second quarter cash used in operating activities of $11.2 million. We expect the cash, cash equivalents and marketable securities at June 30, 2020 are sufficient to meet estimated working capital requirements and fund planned operations through 2023. At June 30, 2020 we had 39.1 million shares outstanding.

I will now turn the call over to Anthony to close.

--------------------------------------------------------------------------------

Anthony S. Marucci, Celldex Therapeutics, Inc. - Founder, President, CEO & Director [6]

--------------------------------------------------------------------------------

Thank you, Sam and thank you all for joining us today. To recap, as always, we remain focused on the successful development of our clinical programs.

We look forward to initiating the 2 Phase 1B studies of CDX-0159 this fall and the Phase 1 study of CDX-527 and the CDX-1140 expansion cohort later this year, followed by the third study of CDX-0159 in an additional mast cell indication next summer.

For data readouts, we plan to present data update for the CDX-1140 program later this year. in 2021, we anticipate data from the CDX-0159 study in chronic inducible urticaria in the first quarter, data from CDX-527 in the first half and the data from CDX-0159 study and the chronic spontaneous urticaria study in the second half of the year.

I would also anticipate data from the CDX-1140 combination with KEYTRUDA and other expansion cohorts in 2021. As Sam said, we are well capitalized to complete the studies necessary to reach these milestones and and for that, I'd like to thank the investors that participated in our recent financing. We look forward to keeping you all up to date as we continue our progress on these programs.

With that review, we will open the floor to questions. operator?

================================================================================

Questions and Answers

--------------------------------------------------------------------------------

Operator [1]

--------------------------------------------------------------------------------

Thank you. We begin our question and answer session. (Operator Instructions) and our first question comes from Kristen Kluska of Cantor Fitzgerald.

.

--------------------------------------------------------------------------------

Kristen Brianne Kluska, Cantor Fitzgerald & Co., Research Division - Analyst [2]

--------------------------------------------------------------------------------

Hi, everyone. Thanks for taking my questions and congrats on the great progress that you've made over this past quarter. So the first question is for the CDS-0159 program, given that some of these patients are likely to have co-morbidities, I'm wondering if you might think these are worth evaluating in the background in either or both the Phase 1B and Phase 2 studies to provide any early proof of effect? Given these indications could also be mast cell driven.

--------------------------------------------------------------------------------

Anthony S. Marucci, Celldex Therapeutics, Inc. - Founder, President, CEO & Director [3]

--------------------------------------------------------------------------------

Sure, Kristen. Thanks. This is Anthony, I'll have Diane answer that question.

--------------------------------------------------------------------------------

Diane C. Young, [4]

--------------------------------------------------------------------------------

Yes. So that's an excellent- very good point Kristen. There is a lot of overlap and other conditions that overlap that there may also be impacted. So that is our intention to -- even in those early studies to try to capture what other co-morbidities the patients have and to try to with that response in some way.

--------------------------------------------------------------------------------

Kristen Brianne Kluska, Cantor Fitzgerald & Co., Research Division - Analyst [5]

--------------------------------------------------------------------------------

Great, thank you. And then as it relates to choosing the third mast cells you have an indicating studies in the summer of next year, I wanted to ask if you think the results from the CINDU trial in the first quarter of next year will in any way help determine which one you ultimately choose as the third indication.

--------------------------------------------------------------------------------

Diane C. Young, [6]

--------------------------------------------------------------------------------

Yes. So I think we'll definitely take the data from the CINDU study that's going to give us information about how we're impacting mast cells and some ideas of dose and duration of clinical effect. So I think that will definitely help to inform what we do next year.

--------------------------------------------------------------------------------

More here:
Edited Transcript of CLDX.OQ earnings conference call or presentation 6-Aug-20 8:30pm GMT - Yahoo Finance

Gene Therapy Market By Industry Type, By Brand And Major Players 2020-2027 – Market Research Posts

The report covers the forecast and analysis of the gene therapy market on a global and regional level. The study provides historical data from 2015 to 2018 along with a forecast from 2019 to 2027 based on revenue (USD Million). The study includes drivers and restraints of the gene therapy market along with the impact they have on the demand over the forecast period. Additionally, the report includes the study of opportunities available in the gene therapy market on a global level.

In order to give the users of this report a comprehensive view of the gene therapy market, we have included a competitive landscape and an analysis of Porters Five Forces model for the market. The study encompasses a market attractiveness analysis, wherein all the segments are bench marked based on their market size, growth rate, and general attractiveness.

Download Sample of This Strategic Report:https://www.kennethresearch.com/sample-request-10170423

The report provides company market share analysis to give a broader overview of the key players in the market. In addition, the report also covers key strategic developments of the market including acquisitions & mergers, new service launches, agreements, partnerships, collaborations & joint ventures, research & development, and regional expansion of major participants involved in the market on a global and regional basis.

The study provides a decisive view of the gene therapy market by segmenting the market based on the type, vector type, therapy area, and regions. All the segments have been analyzed based on present and future trends and the market is estimated from 2019 to 2027. The regional segmentation includes the current and forecast demand for North America, Europe, Asia Pacific, Latin America, and the Middle East and Africa.

Gene therapy is utilized for treating neurodegenerative disorders like Alzheimer, amyotrophic lateral sclerosis, and spinal muscular atrophy. Gene therapy is one of the key treatment kinds that will propel the market growth over the forecast period. Moreover, gene therapy also finds lucrative applications in precision medicine. In addition to this, a rise in the occurrence of cancer is prompting the demand to treat the disease through gene therapy.

Request For Full Report:https://www.kennethresearch.com/sample-request-10170423

Based on the type, the market can be segregated into Germ Line Gene Therapy and Somatic Gene Therapy. In terms of vector type, the gene therapy industry can be divided into Viral Vectors, Non-Viral Vectors, and Human Artificial Chromosome. On the basis of therapy area, the market for gene therapy can be classified into Cancer, Neurological Diseases, Infectious Diseases, Genetic Disorders, Rheumatoid Arthritis, and Others.

The key players included in this market are Advanced Cell & Gene Therapy, Audentes Therapeutics, Benitec Biopharma, Biogen, Blubird Bio, Inc., Bristol-Myers Squibb Company, CHIESI Farmaceutici SPA, Eurofins Scientific, Geneta Science, Genzyme Corporation, Gilead, GlaxoSmithKline PLC, Human Stem Cells institute, Novartis AG, Orchard Therapeutics, Pfizer Inc., Sangamo therapeutics, Spark therapeutics, and Voyager Therapeutics.

About Kenneth Research

Kenneth Research is a reselling agency providing market research solutions in different verticals such as Automotive and Transportation, Chemicals and Materials, Healthcare, Food & Beverage and Consumer Packaged Goods, Semiconductors, Electronics & ICT, Packaging, and Others. Our portfolio includes set of market research insights such as market sizing and market forecasting, market share analysis and key positioning of the players (manufacturers, deals and distributors, etc), understanding the competitive landscape and their business at a ground level and many more. Our research experts deliver the offerings efficiently and effectively within a stipulated time. The market study provided by Kenneth Research helps the Industry veterans/investors to think and to act wisely in their overall strategy formulation

Contact Us

Kenneth Research

Email : [emailprotected]

Phone: +1 313 462 0609

Read more:
Gene Therapy Market By Industry Type, By Brand And Major Players 2020-2027 - Market Research Posts

CAREDX : MANAGEMENT’S DISCUSSION AND ANALYSIS OF FINANCIAL CONDITION AND RESULTS OF OPERATIONS (form 10-Q) – marketscreener.com

The following discussion and analysis of our financial condition and results ofoperations should be read together with the unaudited condensed consolidatedfinancial statements and related notes included elsewhere in Item 1 of Part I ofthis Quarterly Report on Form 10-Q and with the audited consolidated financialstatements and the related notes included in our Annual Report on Form 10-K forthe fiscal year ended December 31, 2019, filed with the Securities and ExchangeCommission, or the SEC, on February 28, 2020.SPECIAL NOTE REGARDING FORWARD-LOOKING STATEMENTSThis Quarterly Report on Form 10-Q contains forward-looking statements withinthe meaning of Section 27A of the Securities Act of 1933, as amended, andSection 21E of the Securities Exchange Act of 1934, as amended. All statementscontained in this Quarterly Report on Form 10-Q other than statements ofhistorical fact, including statements regarding our future results of operationsand financial position, our business strategy and plans, and our objectives forfuture operations, are forward-looking statements. The words "believe," "may,""will," "potentially," "estimate," "continue," "anticipate," "intend," "could,""should," "would," "project," "plan," "target," "contemplate," "predict,""expect" and the negative and plural forms of these words and similarexpressions are intended to identify forward-looking statements.These forward-looking statements may include, but are not limited to, statementsconcerning the following:the potential impact to our business, revenue, financial condition andemployees, including disruptions to our testing services, laboratories, clinicaltrials, supply chain and operations, due to the COVID-19 global pandemic;our ability to take advantage of opportunities under the Coronavirus Aid,Relief, and Economic Security Act, or the CARES Act, and the potential impact ofthe CARES Act on our business, results of operations, financial condition orliquidity;our ability to generate revenue and increase the commercial success of ourcurrent and future testing services, products and digital solutions;our ability to obtain, maintain and expand reimbursement coverage from payersfor our current and other future testing services, if any;our plans and ability to continue updating our testing services, products anddigital solutions to maintain our leading position in transplantations;the outcome or success of our clinical trial collaborations and registrystudies; including Kidney Allograft Outcomes AlloSure Registry, or K-OAR, theOutcomes of KidneyCare on Renal Allografts registry study, or OKRA, and theSurveillance HeartCare Outcomes Registry, or SHORE;the favorable review of our testing services and product offerings, and ourfuture solutions, if any, in peer-reviewed publications;our ability to obtain additional financing on terms favorable to us, or at all;our anticipated cash needs and our anticipated uses of our funds, including ourestimates regarding operating expenses and capital requirements;anticipated trends and challenges in our business and the markets in which weoperate;our dependence on certain of our suppliers, service providers and otherdistribution partners; 25-------------------------------------------------------------------------------- Table of Contentsdisruptions to our business, including disruptions at our laboratories andmanufacturing facilities;our ability to retain key members of our management team;our ability to make successful acquisitions or investments and to manage theintegration of such acquisitions or investments;our ability to expand internationally;our compliance with federal, state and foreign regulatory requirements;our ability to protect and enforce our intellectual property rights, ourstrategies regarding filing additional patent applications to strengthen ourintellectual property rights, and our ability to defend against intellectualproperty claims that may be brought against us;our ability to successfully assert, defend against or settle any litigationbrought by or against us or other legal matters or disputes; andour ability to comply with the requirements of being a public company.These forward-looking statements are subject to a number of risks, uncertaintiesand assumptions, including those described in the section entitled "RiskFactors" in this Quarterly Report on Form 10-Q and in our Annual Report on Form10-K for the fiscal year ended December 31, 2019, filed with the SEC onFebruary 28, 2020. Moreover, we operate in a very competitive and rapidlychanging environment, and new risks emerge from time to time. It is not possiblefor our management to predict all risks, nor can we assess the impact of allfactors on our business or the extent to which any factor, or combination offactors, may cause actual results to differ materially and adversely from thosecontained in any forward-looking statements we may make. In light of theserisks, uncertainties and assumptions, the forward-looking events andcircumstances discussed in this report may not occur and actual results coulddiffer materially and adversely from those anticipated or implied in theforward-looking statements.You should not rely upon forward-looking statements as predictions of futureevents. Although we believe that the expectations reflected in theforward-looking statements are reasonable, we cannot guarantee that the futureresults, levels of activity, performance or events and circumstances reflectedin the forward-looking statements will be achieved or occur. Moreover, neitherwe nor any other person assumes responsibility for the accuracy and completenessof the forward-looking statements. Except as required by law, we undertake noobligation to update publicly any forward-looking statements for any reasonafter the date of this report to conform these statements to actual results orto changes in our expectations.You should read this Quarterly Report on Form 10-Q and the documents that wereference in this Quarterly Report on Form 10-Q and have filed with the SEC asexhibits to this Quarterly Report on Form 10-Q with the understanding that ouractual future results, levels of activity, performance and events andcircumstances may be materially different from what we expect. We qualify allforward-looking statements by these cautionary statements.Overview and Recent HighlightsCareDx, Inc. (collectively, the "Company", "we", "us" and "our") is a leadingprecision medicine company focused on the discovery, development andcommercialization of clinically differentiated, high-value diagnostic solutionsfor transplant patients and caregivers. We offer testing services, products, anddigital healthcare solutions along the pre- and post-transplant patient journey,and we are a leading provider of genomics-based information for transplantpatients.Highlights for the Three Months Ended June 30, 2020 and Recent HighlightsAchieved total revenue of $41.8 million for the three months ended June 30,2020, increasing 33% year-over-yearProvided over 17,100 AlloSure Kidney and AlloMap Heart patient results, withover 40% originating from RemoTraC and mobile phlebotomyRecorded first-ever AlloCell revenue from a cell therapy partnershipCompleted successful public offering raising $134.6 million in net proceeds,increasing cash and cash equivalents to approximately $211.4 millionTesting ServicesHeart 26-------------------------------------------------------------------------------- Table of ContentsAlloMap Heart is a gene expression test that helps clinicians monitor andidentify heart transplant recipients with stable graft function who have a lowprobability of moderate-to-severe acute cellular rejection. Since 2008, we havesought to expand the adoption and utilization of our AlloMap Heart solutionthrough ongoing studies to substantiate the clinical utility and actionability,secure positive reimbursement decisions from large private and public payers,develop and enhance our relationships with key members of the transplantcommunity, including opinion leaders at major transplant centers, and exploreopportunities and technologies for the development of additional solutions forpost-transplant surveillance.We believe the use of AlloMap Heart, in conjunction with other clinicalindicators, can help healthcare providers and their patients better managelong-term care following a heart transplant, can improve patient care by helpinghealthcare providers avoid the use of unnecessary, invasive surveillancebiopsies and may help to determine the appropriate dosage levels ofimmunosuppressants. In 2008, AlloMap Heart received 510(k) clearance from theU.S. Food and Drug Administration for marketing and sale as a test to aid in theidentification of heart transplant recipients, who have a low probability ofmoderate/severe acute cellular rejection at the time of testing, in conjunctionwith standard clinical assessment.AlloMap Heart has been a covered service for Medicare beneficiaries sinceJanuary 1, 2006. The Medicare reimbursement rate for AlloMap Heart is currently$3,240. AlloMap Heart has also received positive coverage decisions forreimbursement from many of the largest U.S. private payers, including Aetna,Anthem, Cigna, Health Care Services Corporation, or HCSC, Humana, KaiserFoundation Health Plan, Inc. and UnitedHealthcare.We have also successfully completed a number of landmark clinical trials in thetransplant field demonstrating the clinical utility of AlloMap Heart forsurveillance of heart transplant recipients. We initially established theanalytical and clinical validity of AlloMap Heart on the basis of our CardiacAllograft Rejection Gene Expression Observational (Deng, M. et al., Am JTransplantation 2006), or CARGO, study, which was published in the AmericanJournal of Transplantation. A subsequent clinical utility trial, InvasiveMonitoring Attenuation through Gene Expression (Pham MX et al., N. Eng. J. Med.,2010), or IMAGE, published in The New England Journal of Medicine, demonstratedthat clinical outcomes in recipients managed with AlloMap Heart surveillancewere equivalent (non-inferior) to outcomes in recipients managed withbiopsies. The results of our clinical trials have also been presented at majormedical society congresses. AlloMap Heart is now recommended as part of theInternational Society for Heart and Lung Transplantation, or ISHLT, guidelines.HeartCareHeartCare includes the gene expression profiling technology of AlloMap Heartwith the dd-cfDNA analysis of AlloSure Heart in one surveillance solution. Anapproach to surveillance using HeartCare provides information from twocomplementary measures: (i) AlloMap Heart - a measure of immune activation, and(ii) AlloSure Heart - a measure of graft injury.Clinical validation data from the Donor-Derived Cell-Free DNA-Outcomes AlloMapRegistry (NCT02178943), or D-OAR, was published in American Journal ofTransplant, or AJT, in 2019. D-OAR was an observational, prospective,multicenter study to characterize the AlloSure-Heart dd-cfDNA in a routine,clinical surveillance setting with heart transplant recipients. The D-OAR studywas designed to validate that plasma levels of AlloSure-Heart dd-cfDNA candiscriminate acute rejection from no rejection, as determined by endomyocardialbiopsy criteria.HeartCare provides robust information about distinct biological processes, suchas immune quiescence, active injury, Acute Cellular Rejection, or ACR, andAntibody Mediated Rejection. In September 2018, we initiated the SHORE study.SHORE is a prospective, multi-center, observational, registry of patientsreceiving HeartCare for surveillance. Patients enrolled in SHORE will befollowed for 5 years with collection of clinical data and assessment of 5-yearoutcomes.In August 2019, AlloSure Heart received a positive draft Local CoverageDetermination for Medicare coverage. We have not yet made any applications toprivate payers for reimbursement coverage of AlloSure Heart.KidneyAlloSure Kidney, our transplant surveillance solution, which was commerciallylaunched in October 2017, is our donor-derived cell-free DNA, or dd-cfDNA,offering built on a Next Generation Sequencing, or NGS, platform. Intransplantation, 109 papers from 55 studies globally have shown the value ofdd-cfDNA in the management of solid organ transplantation. AlloSure allowssequencing of DNA and RNA much more quickly than the previously used Sangersequencing. AlloSure is able to discriminate dd-cfDNA from recipient-cell-freeDNA, targeting polymorphisms between donor and recipient. This single-nucleotidepolymorphism, or SNPs, approach across all the somatic chromosomes isspecifically designed for transplantation, allowing a scalable, high-qualitytest to differentiate dd-cfDNA.AlloSure Kidney has received positive coverage decisions for reimbursement fromMedicare. The Medicare reimbursement rate for AlloSure Kidney is $2,841.AlloSure Kidney has also received positive coverage decisions from BCBS SouthCarolina and BCBS Kansas City, and is reimbursed by other private payers on acase-by-case basis. 27-------------------------------------------------------------------------------- Table of ContentsMultiple studies have demonstrated that significant allograft injury can occurin the absence of changes in serum creatinine. Thus, clinicians have limitedability to detect injury early and intervene to prevent long term damage usingthis marker. While histologic analysis of the allograft biopsy specimen remainsthe standard method used to assess injury and differentiate rejection from otherinjury in kidney transplants, as an invasive test with complications, repetitivebiopsies are not well tolerated. AlloSure provides a non-invasive test,assessing allograft injury that enables more frequent, quantitative and saferassessment of allograft rejection and injury status. Beyond allograft rejection,the assessment of molecular inflammation has shown further utility in theassessment of proteinuria, formation of De Novo donor specific antibodies, orDSAs, and also as a surrogate predictive measure of estimated glomerularfiltration rate, or eGFR, decline. Monitoring of graft injury through AlloSureallows clinicians to optimize allograft biopsies, identify allograft injury andguide immunosuppression management more accurately.Since the analytical validation paper in the Journal of Molecular Diagnostics in2016 before the commercial launch of AlloSure Kidney, an increasing body ofevidence supports the use of AlloSure dd-cfDNA in the assessment andsurveillance of kidney transplants. Bloom et al evaluated 102 kidney recipientsand demonstrated that dd-cfDNA levels could discriminate accurately andnon-invasively distinguish rejection from other types of graft injury. Incontrast, serum creatinine has area under the curve, or AUC, of 50%, showing nosignificant difference between patients with and without rejection. Multiplepublications and abstracts have shown AlloSure's value in the management of BKviremia, as well as numerous pathologies that cause molecular inflammation andinjury such as DSAs and eGFR decline. Most recently its utility in theassessment of T-cell mediated rejection (TCMR) 1A and borderline rejection hasalso been published in the AJT.The prospective multicenter trial: Kidney Allograft Outcomes AlloSure KidneyRegistry, or the K-OAR study, is currently ongoing and has enrolled over 1,600patients, with plans to survey patients with AlloSure for 3 years and providefurther clinical utility of AlloSure Kidney in the surveillance of kidneytransplant recipients.KidneyCareKidneyCare combines the dd-cfDNA analysis of AlloSure Kidney with the geneexpression profiling technology of AlloMap Kidney and the predictive artificialintelligence technology of KidneyCare iBox in one surveillance solution. We havenot yet made any applications to payers for reimbursement coverage of AlloMapKidney or KidneyCare iBox.In September 2019, we announced the enrollment of the first patient in the OKRAstudy, which is an extension of the K-OAR study. OKRA is a prospective,multi-center, observational registry of patients receiving KidneyCare forsurveillance. Combined with K-OAR, 4,000 patients will be enrolled into thestudy.LungIn February 2019, AlloSure Lung became available for lung transplant patientsthrough a compassionate use program while the test is undergoing furtherstudies. AlloSure Lung applies proprietary NGS technology to measure dd-cfDNAfrom the donor lung in the recipient bloodstream to monitor graft injury. Wehave not yet made any applications to payers for reimbursement coverage ofAlloSure Lung.Cellular TherapyIn April 2020, we initiated a research partnership for AlloCell, a surveillancesolution that monitors the level of engraftment and persistence of allogeneiccells for patients who have received cell therapy transplants. AlloCell willinitially be commercialized through collaborative research agreements withbiopharma companies developing cell therapies.ProductsWe develop, manufacture, market and sell products that increase the chance ofsuccessful transplants by facilitating a better match between a solid organ orstem cell donor and a recipient, and help to provide post-transplantsurveillance of these recipients.QTYPE enables Human Leukocyte Antigen or HLA typing at a low to intermediateresolution for samples that require a fast turn-around-time and uses real-timepolymerase chain reaction, or PCR, methodology. Olerup SSP is used to type HLAalleles based on the sequence specific primer, or SSP, technology. Olerup SBT isa complete product range for sequence-based typing of HLA alleles.On May 4, 2018, we entered into a license agreement with Illumina, Inc., or theIllumina Agreement, which provides us with worldwide distribution, developmentand commercialization rights to Illumina's NGS products and technologies for usein transplantation diagnostic testing.On June 1, 2018, we became the exclusive worldwide distributor of Illumina'sTruSight HLA product line. TruSight HLA is a high-resolution solution that usesNGS methodology. In addition, we were granted the exclusive right to develop andcommercialize other NGS product lines in the field of bone marrow and solidorgan transplantation on diagnostic testing. These 28-------------------------------------------------------------------------------- Table of ContentsNGS products include: AlloSeq Tx, a high-resolution HLA typing solution, AlloSeqcfDNA, our surveillance solution designed to measure dd-cfDNA in blood to detectactive rejection in transplant recipients, and AlloSeq HCT, a NGS solution forchimerism testing for stem cell transplant recipients.In September 2019, we commercially launched AlloSeq cfDNA, our surveillancesolution designed to measure dd-cfDNA in blood to detect active rejection intransplant recipients, and we received CE mark approval on January 10, 2020. Ourability to increase the clinical uptake for AlloSeq cfDNA will be a result ofmultiple factors including local clinical education, customer lab technicalproficiency and levels of country-specific reimbursement.Also in September 2019, we commercially launched AlloSeq Tx, the first of itskind NGS high-resolution HLA typing solution utilizing hybrid capturetechnology. This technology enables the most comprehensive sequencing, coveringmore of the HLA genes than current solutions and adding coverage of non-HLAgenes that may impact transplant patient matching and management. AlloSeq Tx hassimple NGS workflow, with a single tube for processing and steps to reduceerrors. AlloSeq Tx 17 received CE mark approval on May 15, 2020.In June 2020, we commercially launched AlloSeq HCT, a NGS solution for chimerismtesting for stem cell transplant recipients. This technology can provide bettersensitivity and data analysis compared to current solutions on the market.DigitalIn 2019, we began providing digital solutions to transplant centers followingthe acquisition of Ottr Complete Transplant Management, or OttrCare, andXynManagement, Inc., or XynManagement.On May 7, 2019, we acquired 100% of the outstanding common stock of OttrCare.OttrCare was formed in 1993 and is a leading provider of transplant patienttracking software, or the Ottr software, which provides comprehensive solutionsfor transplant patient management. The Ottr software enables integration withelectronic medical records, systems, including Cerner and Epic, providingpatient surveillance management tools and outcomes data to transplant centers.On August 26, 2019, we acquired 100% of the outstanding common stock ofXynManagement. XynManagement provides two unique solutions, XynQAPI software, orXynQAPI, and Waitlist Management. XynQAPI simplifies transplant quality trackingand Scientific Registry of Transplant Recipients, or SRTR, reporting. WaitlistManagement includes a team of transplant assistants who maintain regular contactwith patients on the waitlist to help prepare for their transplant and maintaineligibility.COVID-19 ImpactIn the final weeks of March and during April 2020, with hospitals increasinglycaring for COVID-19 patients, hospital administrators chose to limit or evendefer, non-emergency procedures. Immunosuppressed transplant patients eitherself-prescribed or were asked to avoid transplant centers and caregiver visitsto reduce the risk of contracting COVID-19. As a result, with transplantsurveillance visits down, we experienced a slowdown in testing services volumesin the final weeks of March and during April 2020. As a response to the COVID-19pandemic, and to enable immune-compromised transplant patients to continue tohave their blood drawn, in late March 2020 we launched RemoTraC, a remotehome-based blood draw solution using mobile phlebotomy for AlloSure and AlloMapsurveillance tests, as well as for other standard monitoring tests. To date,more than 150 transplant centers can offer RemoTraC to their patients and over4,000 kidney, heart, and lung transplant patients have enrolled. Based onexisting and new relationships with partners, we have established a nationwidenetwork of more than 10,000 mobile phlebotomists. Following the introduction ofRemoTraC and with the easing of stay-at-home restrictions and the opening up ofmany hospitals to non-COVID-19 patients, our testing services volumes returnedto levels consistent with those experienced immediately prior to the impact ofCOVID-19, and volumes continued to be at or above those levels throughout May2020 and June 2020. However, our product business experienced a reduction inforecasted sales volume throughout the second quarter 2020, as we were unable toundertake onsite discussions and demonstrations of our recently launched NGSproducts, including AlloSeq Tx 17, which was awarded CE mark approval in May2020.We are maintaining our testing, manufacturing, and distribution facilities whileimplementing specific protocols to reduce contact among our employees. In areaswhere COVID-19 impacts healthcare operations, our field-based sales and clinicalsupport teams are supporting providers through telephone and online platforms.To reduce the risk to employees and their families from potential exposure toCOVID-19, most of our corporate employees have been asked to work from home. Wehave also restricted non-essential business travel to protect the health andsafety of its employees, patients, and customers. In addition, we have created aCOVID-19 task force that is responsible for crisis decision making, employeecommunications, enforcing pre-arrival temperature checking, daily healthcheck-ins and enhanced safety training/protocols in our offices for employeesthat cannot work from home.Due to COVID-19, quarantines, shelter-in-place and similar government orders, orthe perception that such orders, shutdowns or other restrictions on the conductof business operations could occur or could impact personnel at third-partysuppliers in the United States and other countries, or the availability or costof materials, there may be disruptions in our supply chain. Any 29-------------------------------------------------------------------------------- Table of Contentsmanufacturing supply interruption of materials could adversely affect ourability to conduct ongoing and future research and testing activities.In addition, our clinical studies may be affected by the COVID-19 pandemic.Clinical site initiation and patient enrollment may be delayed due toprioritization of hospital resources toward the COVID-19 pandemic. Some patientsmay not be able to comply with clinical study protocols if quarantines impedepatient movement or interrupt healthcare services. Similarly, the ability torecruit and retain patients and principal investigators and site staff who, ashealthcare providers, may have heightened exposure to COVID-19, may adverselyimpact our clinical trial operations.Financial Operations OverviewRevenueWe derive our revenue from testing services, products sales and digital andother revenues. Revenue is recorded considering a five-step revenue recognitionmodel that includes identifying the contract with a customer, identifying theperformance obligations in the contract, determining the transaction price,allocating the transaction price to the performance obligations and recognizingrevenue when, or as, an entity satisfies a performance obligation.Testing Services RevenueOur testing services revenue is derived from AlloSure Kidney and AlloMap Hearttests, which represented 87% and 84% of our total revenues for the three and sixmonths ended June 30, 2020, respectively, and 82% of our total revenues for eachof the three and six months ended June 30, 2019. Our testing services revenuedepends on a number of factors, including (i) the number of tests performed;(ii) establishment of coverage policies by third-party insurers and governmentpayers; (iii) our ability to collect from payers with whom we do not havepositive coverage determination, which often requires that we pursue acase-by-case appeals process; (iv) our ability to recognize revenues on testsbilled prior to the establishment of reimbursement policies, contracts orpayment histories; (v) our ability to expand into markets outside of the UnitedStates; and (vi) how quickly we can successfully commercialize new productofferings.We currently market testing services to healthcare providers through our directsales force that targets transplant centers and their physicians, coordinatorsand nurse practitioners. The healthcare providers that order the tests and onwhose behalf we provide our testing services are generally not responsible forthe payment of these services. Amounts received by us vary from payer to payerbased on each payer's internal coverage practices and policies. We generallybill third-party payers upon delivery of a test result report to the orderingphysician. As such, we take the assignment of benefits and the risk ofcollection from the third-party payer and individual patients.In April 2020, we announced our first biopharma research partnership forAlloCell, a surveillance solution that monitors the level of engraftment andpersistence of allogeneic cells for patients who have received cell therapytransplants. AlloCell will initially be commercialized through collaborativeresearch agreements with biopharma companies developing cell therapies.Product RevenueOur product revenue is derived primarily from sales of Olerup SSP, QTYPE,TruSight and AlloSeq Tx products. Product revenue represented 8% and 10% oftotal revenue for the three and six months ended June 30, 2020, respectively,and 15% and 16% of total revenue for the three and six months ended June 30,2019, respectively. We recognize product revenue from the sale of products toend-users, distributors and strategic partners when all revenue recognitioncriteria are satisfied. We generally have a contract or a purchase order from acustomer with the specified required terms of order, including the number ofproducts ordered. Transaction prices are determinable and products are deliveredand risk of loss passed to the customer upon either shipping or delivery, as perthe terms of the agreement. There are no further performance obligations relatedto a contract and revenue is recognized at the point of delivery consistent withthe terms of the contract or purchase order.Digital and Other RevenueOur digital and other revenue is mainly derived from sales of our Ottr softwareand XynQAPI licenses and services and other licensing agreements. Digital andother revenue represented 5% and 6% of total revenue for the three and sixmonths ended June 30, 2020, respectively, and 4% and 2% of total revenue for thethree and six months ended June 30, 2019, respectively.Critical Accounting Policies and Significant Judgments and EstimatesOur management's discussion and analysis of our financial condition and resultsof operations is based on our unaudited condensed consolidated financialstatements, which have been prepared in accordance with United States generallyaccepted accounting principles. The preparation of these unaudited condensedconsolidated financial statements requires us to make estimates and assumptionsthat affect the reported amounts of assets and liabilities and the disclosure ofcontingent assets and liabilities at the date of the unaudited condensedconsolidated financial statements, as well as the reported revenue generated 30-------------------------------------------------------------------------------- Table of Contentsand expenses incurred during the reporting periods. Our estimates are based onour historical experience and on various other factors that we believe arereasonable under the circumstances, the results of which form the basis formaking judgments about the carrying value of assets and liabilities that are notreadily apparent from other sources. Actual results may differ from theseestimates under different assumptions or conditions.We believe that the following critical accounting policies reflect the moresignificant estimates and assumptions used in the preparation of our financialstatements. We believe the following critical accounting policies are affectedby significant judgments and estimates used in the preparation of our unauditedcondensed consolidated financial statements:Revenue recognition;Business combination;Acquired intangible assets;Impairment of goodwill, intangible assets and other long-lived assets; andCommon stock warrant liability.There were no material changes in the matters for which we make criticalaccounting estimates in the preparation of our unaudited condensed consolidatedfinancial statements during the three and six months ended June 30, 2020 ascompared to those disclosed in Management's Discussion and Analysis of FinancialCondition and Results of Operations included in our annual report on Form 10-Kfor the year ended December 31, 2019, except that there is no derivativeliability outstanding as of December 31, 2019 and June 30, 2020 and thedetermination of the estimated present value of lease payments using ourincremental borrowing rate as discussed in Note 2, Summary of SignificantAccounting Policies, in the unaudited condensed consolidated financialstatements included elsewhere in this Quarterly Report on Form 10-Q.Recently Issued Accounting StandardsRefer to Note 2, Summary of Significant Accounting Policies - Recent AccountingPronouncements, to the unaudited condensed consolidated financial statementsincluded elsewhere in this Quarterly Report on Form 10-Q for a description ofrecently issued accounting pronouncements, including the expected dates ofadoption and estimated effects on our results of operations, financial positionand cash flows. 31-------------------------------------------------------------------------------- Table of ContentsResults of OperationsComparison of the Three Months Ended June 30, 2020 and 2019(In thousands) Three Months Ended June 30, 2020 2019 ChangeRevenue:Testing services revenue $ 36,293$ 25,677$ 10,616Product revenue 3,291 4,593 (1,302)Digital and other revenue 2,217 1,184 1,033Total revenue 41,801 31,454 10,347Cost of revenue 15,025 11,512 3,513Gross profit 26,776 19,942 6,834Operating expenses:Research and development 13,129 7,630 5,499Sales and marketing 12,134 10,644 1,490General and administrative 12,316 8,512 3,804Total operating expenses 37,579 26,786 10,793Loss from operations (10,803) (6,844) (3,959)Other income (expense):Interest income, net 21 300 (279)

Change in estimated fair value of common stock warrant liability

Change in estimated fair value of common stock warrant liability

Effect of exchange rate changes on cash, cash equivalents and restricted cash

--------------------------------------------------------------------------------

Edgar Online, source Glimpses

Continued here:
CAREDX : MANAGEMENT'S DISCUSSION AND ANALYSIS OF FINANCIAL CONDITION AND RESULTS OF OPERATIONS (form 10-Q) - marketscreener.com

Tooth Regeneration Market Size -Future of growth, Top Capital Facts, Global Industry Analysis, Share, Growth, Trends and Forecast 2020 – 2025 – Market…

Detailed Analysis & SWOT analysis, Tooth Regeneration Market Trends 2020, Tooth Regeneration Market Growth 2020, Tooth Regeneration Industry Share 2020, Tooth Regeneration Industry Size, Tooth Regeneration Market Research, Tooth Regeneration Market Analysis,

GlobalTooth Regeneration Marketreport tends to a portion of the major and one of a kind parts of the market. Further report makes reference to mechanical norms based on planned degree and industry measurement and uncovers. Based on notable information, showcase size has been determined as far as income from base year 2020 to 2025.

The global Tooth Regeneration market is anticipated to rise at a considerable rate during the forecast period, between 2020 and 2024.

Get a sample copy of the Tooth Regeneration market report 2020

Tooth Regeneration Industry Outlook 2020:->

Tooth regeneration is a stem cell based regenerative medicine procedure in the field of tissue engineering and stem cell biology to replace damaged or lost teeth by regrowing them from autologous stem cells. As a source of the new bioengineered teeth somatic stem cells are collected and reprogrammed to induced pluripotent stem cells which can be placed in the dental lamina directly or placed in a reabsorbable biopolymer in the shape of the new tooth.,

Tooth Regeneration market competition by top Countries manufacturers/ Key player Data Profiled:

And More

Get a Sample PDF of report @https://www.marketgrowthreports.com/enquiry/request-sample/14428634

Scope of the Tooth Regeneration MarketReport:

Tooth Regeneration Market Segment by Type covers:

Tooth Regeneration Market Segment by Applications can be divided into:

Regional analysis covers:

Fill the Pre-Order Enquiry form for the report @https://www.marketgrowthreports.com/enquiry/pre-order-enquiry/14428634

This Tooth Regeneration Market Research/Analysis Report Contains Answers to your following Questions

Key Benefits

And More.

Look into Table of Content of Tooth Regeneration Market Report @https://www.marketgrowthreports.com/TOC/14428634#TOC

The next part also sheds light on the gap between supply and consumption. Apart from the mentioned information,growth rateof Tooth Regeneration market in 2025is also explained.Additionally, type wise and application wise consumptiontables andfiguresof Tooth Regeneration marketare also given.

Buy this report (Price USD 3480 for a single-user license)@https://www.marketgrowthreports.com/purchase/14428634

About 360 Market Updates:

360 Market Updates is the credible source for gaining the market research reports that will exponentially accelerate your business. We are among the leading report resellers in the business world committed towards optimizing your business. The reports we provide are based on a research that covers a magnitude of factors such as technological evolution, economic shifts and a detailed study of market segments.

CONTACT US

Mr. Ajay More

Phone:+14242530807 / + 44 20 3239 8187

Email:[emailprotected]

For More Related Reports Click Here :

Virtual Cards Market Size 2020 : Growth Factors, Business Opportunity,Segmentation and Forecast to 2026 Research Reportby implies ofMarket Growth Reports

LED Materials Market Size 2020 : Growth Factors, Business Opportunity,Segmentation and Forecast to 2026 Research Reportby implies ofMarket Growth Reports

Vulkollan Wheels Market Size and Growth, Sales Revenue, Key Players Analysis, Development Status, Opportunity Assessment and Industry Expansion Strategies 2025 Research ReportthroughMarket Growth Reports

Seamless Steel Tube Market Size, Growth Opportunities, Emerging Technologies, Trends,Growth, Segments, Landscape and Demandby route ofForecast to 2026 Research Reportwith the guide ofMarket Growth Reports

Chocolate Ice Cream Market Size, Growth Opportunities, Emerging Technologies, Trends,Growth, Segments, Landscape and Demandby usingForecast to 2026 Research ReportviaMarket Growth Reports

Read the rest here:
Tooth Regeneration Market Size -Future of growth, Top Capital Facts, Global Industry Analysis, Share, Growth, Trends and Forecast 2020 - 2025 - Market...

Somatic Stem Cells | SpringerLink

Stem cells are found in almost all organisms from the early stages of development to the end of life. There are several types of stem cells and all of them may prove useful for medical research; however, each of the different types has both promise and limitations. Somatic Stem Cells: Methods and Protocols presents selected genetic, molecular, and cellular techniques used in somatic stem cell research and its clinical application. Chapters focus on the isolation, characterization, purity, plasticity, and clinical uses of somatic stem cells from a variety of human and animal tissues. Written in the highly successful Methods in Molecular Biology series format, chapters include introductions to their respective topics, lists of the necessary materials and reagents, step-by-step, readily reproducible laboratory protocols, and key tips on troubleshooting and avoiding known pitfalls.

Through and intuitive, Somatic Stem Cells: Methods and Protocols seeks to provides scientists with the fundamental techniques of stem cell research and its potential application in regenerative medicine.

Somatic Stem cells anticancer therapies embryonic stem cells pluripotent stem cells regenerative medicine stem cell differentiation stem cell proliferation tissue homeostasis

Link:
Somatic Stem Cells | SpringerLink

-Catenin safeguards the ground state of mousepluripotency by strengthening the robustness of the transcriptional apparatus – Science Advances

INTRODUCTION

Pluripotency can be sustained in vitro through culture in specific conditions. Mouse embryonic stem cells (ESCs) in conventional serum/leukemia inhibitory factor (LIF) (SL) medium are considered to exhibit nave, preimplantation-like pluripotency because they contribute to chimeras with relative high efficiency upon blastocyst complementation. Yet, only a proportion of ESCs in SL are truly nave at a given time, and the entire population is highly metastable, periodically switching between nave and early post-implantationlike (formative or partially primed) pluripotent states (1, 2). Culture in serum-free medium with mitogen-activated protein kinase kinase (MEK) and glycogen synthase kinase 3 (GSK3) inhibitors, PD0325901 (PD) and CHIR99021 (CHIR), produces a pluripotent ground state that more closely resembles the preimplantation inner cell mass (35). Addition of LIF to the 2i (2iL) facilitates pluripotency maintenance in the ground state but is not strictly necessary. ESCs in 2iL have lower expression of lineage-associated genes and more homogeneous expression of pluripotency genes than in SL (6, 7). They also display genome-wide DNA hypomethylation, reduced histone 3 lysine-27 trimethylation (H3K27me3) at promoters, and tolerate better the suppression of epigenetic/epitranscriptomic factors than ESCs in SL (6, 810). Overall, this suggests a rewiring of regulatory networks that confers additional robustness in 2iL, but the underlying mechanisms are unclear.

Gene transcription in eukaryotes has a highly regulated progression involving initiation, Pol2 pausing in the vicinity of the promoter, release of paused Pol2, gene body elongation, and termination (11). Recruitment of the Pol2 transcription initiation apparatus and Pol2 pause release are rate-limiting steps. Initiation is orchestrated by sequence-specific transcription factors (e.g., the pluripotency transcription factors), which, through chromatin remodeling, allow the recruitment of the basal transcription machinery including general transcription factors and Pol2. For many mammalian genes, Pol2 then pauses 20 to 60 nucleotides after the transcription start site (TSS), requiring pause release for subsequent productive gene body elongation. Pol2 pause release is mediated by CDK9, the catalytic subunit of the positive transcription elongation factor b (P-TEFb) complex. CDK9 resides in a catalytically inactive complex that is activated by different mechanisms; the bromodomain and extraterminal (BET) family member BRD4 plays a critical role in this process (12, 13). Pol2 pausing and the subsequent pause release represent a mechanism for ensuring potent but quick binary-switchable gene expression but, being a multistep process, could render gene transcription vulnerable to perturbation. Notably, both transcription initiation and Pol2 pause release are required for sustaining high expression levels of genes involved in pluripotency maintenance and proliferation/self-renewal of mouse ESCs in SL (12, 1416), but it was unclear whether transcriptional regulation in ground-state culture conditions has the same essential requirements.

Here, we show that -catenin potentiates the recruitment of coregulatorsincluding BRD4, CDK9, mediator, cohesin, and p300to strengthen pluripotency loci in ESCs in 2iL. This enhances transcription initiation at those loci, compensatorily lowering the dependence on Pol2 pause release for productive gene body elongation. By contrast, cell cyclerelated genes are not bound by -catenin and remain addicted to Pol2 pause release in 2iL, making self-renewal highly sensitive to BRD4/CDK9 suppression in both culture conditions. Our findings explain how pluripotency gene transcription is selectively reinforced in the ground state to protect against exogenous perturbation.

To investigate distinctive transcriptional features of mouse ESCs cultured in SL or 2iL, we performed a short hairpin RNA (shRNA) screen for a panel of transcriptional regulators (fig. S1A). This panel included regulators of Pol2 pause release, histone methyltransferases/demethylases, histone acetyltransferases/deacetylases, histone acetylation readers, and splicing regulators, many of which are known to be necessary for ESC maintenance in SL (table S1). The effect of the knockdown was determined by measuring the expression of the core pluripotency markers Oct4 (Pou5f1), Nanog, and Klf2 by reverse transcription quantitative polymerase chain reaction (RT-qPCR). Suppressing most regulators had a stronger effect in reducing pluripotency genes in SL than 2iL (Fig. 1A). In particular, we noticed that knocking down two relevant mediators of Pol2 pause release, BRD4 and CDK9, was better tolerated in 2iL.

(A) Heat map showing the relative expression of Pou5f1, Nanog, and Klf2 in ESCs in SL or 2iL transduced with shRNA for the indicated genes. (B) RT-qPCR for the indicated genes in ESCs in SL or 2iL transduced with shRNA for Luciferase (shLuc) or two shRNAs for Brd4 (shBrd4#1 and shBrd4#2). Data are the mean values SEM with the indicated significance (P value was calculated using two-tailed unpaired Students t test, also for all subsequent experiments unless otherwise noted). n = 3. (C) Growth curve of ESCs in 2iL transduced with shLuc, shBrd4#1, or shBrd4#2 measured by cell counting in triplicate at passage 1 after transduction. n = 2. A representative experiment is shown. (D) Percentage of cells in different cell cycle phases in ESCs in 2iL transduced with shLuc, shBrd4#1, or shBrd4#2 measured by flow cytometry at passage 1 after transduction (mean values SEM, n = 3). (E) Phase contrast and alkaline phosphatase (AP) activity of ESCs in SL or 2iL treated with vehicle [dimethyl sulfoxide (DMSO)] or JQ1 at the indicated doses. Scale bar, 50 m. (F) As in (E) but shows RT-qPCR result for the indicated genes (mean values SEM, n = 3). (G) RT-qPCR for the indicated genes in ESCs in 2iL treated with DMSO or JQ1 at the indicated doses (mean values SEM, n = 3). (H) As in (C) but ESCs were treated with DMSO or JQ1 at the indicated doses for passage 0 (P0) or passage 1 (P1). n = 2. A representative experiment is shown. (I) As in (D) but ESCs were treated with DMSO or JQ1 at the indicated doses (mean values SEM, n = 3). (J) Heat map showing the fold change of pluripotency genes and cell cycle genes measured by RNA sequencing (RNA-seq) in ESCs in SL or 2iL treated with DMSO or 100 nM JQ1. *P < 0.05,**P < 0.01, ***P < 0.001.

We first focused on BRD4 because we and others have reported that it is a master regulator of ESC pluripotency/self-renewal (in SL) and early embryonic development (12, 14, 15). Basal BRD4 expression tested by Western blotting was comparable in SL and 2iL (fig. S1B). We repeated the Brd4 knockdown in both conditions and confirmed that it was effective in reducing mRNA and protein expression (Fig. 1B and fig. S1C). In contrast to SL, ESC colonies in 2iL transduced with Brd4 shRNA remained domed and compact, as well as alkaline phosphatase (AP) positive, even after several passages as single cells (fig. S1, D and E). Likewise, pluripotency genes, measured by RT-qPCR, exhibited little change or up-regulation in 2iL compared to SL (Fig. 1B and fig. S1F), but we observed reduced proliferation in both conditions (albeit more obvious in SL) (Fig. 1C and fig. S1D). This was associated with a significant increase in the number of cells in the G0-G1 phase of the cell cycle (Fig. 1D). Analysis of chromatin immunoprecipitationsequencing (ChIP-seq) for BRD4 showed a similar widespread binding pattern in SL and 2iL (fig. S1, G and H). We then validated the differential effects of Brd4 knockdown in SL and 2iL using two additional ESC lines and two more batches of ESC-qualified serum from different vendors (fig. S2, A to E). These results demonstrated that BRD4 is less required for preserving pluripotency in 2iL than SL but remains necessary for self-renewal (i.e., robust proliferative expansion in vitro) under both conditions.

To further verify the differential sensitivity of pluripotency characteristics to BRD4 suppression in ESCs cultured in SL and 2iL, we used JQ1, a well-known BET inhibitor that binds to the two BRD4 bromodomains to prevent their interaction with acetylated histones (17). At lower doses (100 and 200 nM) for 60 hours, JQ1 notably affected colony morphology, AP activity, and pluripotency gene expression in SL but had little effect in 2iL (Fig. 1, E and F). ESCs in 2iL remained competent for teratoma and chimera formation with 100 nM JQ1 (fig. S3, A and B). However, at higher doses (500 nM and above), pluripotency characteristics were also notoriously affected in 2iL (Fig. 1, E and G), especially upon passage as single cells (fig. S3C). Likewise, JQ1 reduced proliferation in 2iL, although at lower doses, this only became prominent after passaging as single cells (Fig. 1H). This was paralleled by an increase in the percentage of cells in G0-G1 and in apoptosis (Fig. 1I and fig. S3D). RNA sequencing (RNA-seq) confirmed the differential effect of 100 nM JQ1 on pluripotency in SL and 2iL and also showed the down-regulation of cell cycle genes (Fig. 1J and table S2). We confirmed that low doses of JQ1 impair pluripotency in SL but not 2iL using two additional ESC lines and two batches of ESC-qualified serum (fig. S3, E to G).

The experiments with JQ1 suggested that a certain level of BRD4 is necessary for maintaining pluripotency in 2iL. To exclude the possibility that higher doses impair pluripotency characteristics in 2iL through off-target effects, we used an inducible Cre/LoxP system for knocking out Brd4 (fig. S4, A to D). Despite extensive testing, we only obtained heterozygous Brd4fl/ clones in 2iL, which proliferated less and differentiated when changed to SL culture conditions (fig. S4, E and F). We also noticed that, in contrast to wild-type clones, low doses of JQ1 could effectively reduce pluripotency gene expression in heterozygous Brd4 knockout ESCs in 2iL (fig. S4G). We concluded that pluripotency maintenance is more resistant to BRD4 suppression in ESCs in 2iL than in SL, but reducing BRD4 beyond a threshold also affects pluripotency in 2iL.

Pol2 pausing is mediated by pausing factors including DRB sensitivityinducing factor (DSIF) and negative elongation factor (NELF), whereas pause release is triggered through phosphorylation of Pol2 on serine-2 (Ser2P) by CDK9. A major role of BRD4 is to induce Pol2 pause release by activating CDK9 (13), a target that was also identified as less necessary for 2iL in our shRNA screen (see above Fig. 1A). Consistently, analysis of CDK9 ChIP-seq in SL showed notable overlap with BRD4 ChIP-seq in SL or 2iL (Fig. 2A). Likewise, a sizeable proportion of genes down-regulated by JQ1 in SL or 2iL were cobound by BRD4 and CDK9 (Fig. 2B), including many pluripotency (in SL) and cell cycle genes (in SL and 2iL) (Fig. 2, B and C, and fig. S5A). To confirm the differential CDK9 dependence in SL and 2iL, we repeated the knockdown experiments and also used a specific CDK9 inhibitor [LDC000067; (18)]. As with Brd4 knockdown, Cdk9 knockdown severely affected colony morphology, AP activity, and pluripotency gene expression in SL but had no obvious effect in 2iL (Fig. 2, D and E), and this persisted for several passages (fig. S5, B and C). Proliferation and the cell cycle were significantly affected by Cdk9 knockdown in 2iL too (Fig. 2, F and G), although to a lesser extent than in SL (Fig. 2D). These effects were validated using an additional ESC line (fig. S5, D and E). Similarly, 10 M LDC000067 impaired colony morphology, AP activity, and pluripotency gene expression in SL but not in 2iL (even after multiple passages as single cells), but a higher dose had severe consequences in both conditions (Fig. 2, H and I, and fig. S5, F and G). Likewise, LDC000067 reduced cell growth in SL and 2iL, impaired the cell cycle, and enhanced apoptosis significantly (Fig. 2, H and J to L). The consistent phenotypes of suppressing Brd4 and Cdk9 implied that reducing Pol2 pause release at pluripotency genes is better tolerated in 2iL than SL, suggesting a major change in transcriptional control in the two culture conditions.

(A) Venn diagrams showing the overlap between BRD4 bound sites in ESCs in SL or 2iL and CDK9 bound sites. (B) Venn diagrams showing the overlap between genes down-regulated by 100 nM JQ1 in ESCs in SL or 2iL and BRD4/CDK9 cobound genes. (C) Genome views for a BRD4/CDK9 cobound pluripotency gene (Nanog) and a cell cycle gene (Mdm4) in ESCs cultured as indicated. (D) Phase contrast and AP activity of ESCs in SL or 2iL transduced with shLuc or two shRNAs for Cdk9 (shCdk9#1 and shCdk9#2). Scale bar, 50 m. (E) As in (D) but shows RT-qPCR result for the indicated genes (mean values SEM, n = 5). (F) Growth curve of ESCs in 2iL transduced with shLuc, shCdk9#1, or shCdk9#2 measured by cell counting in triplicate at passage 1 after transduction. n = 2. A representative experiment is shown. (G) Percentage of cells in different cell cycle phases in ESCs in 2iL transduced with shLuc, shCdk9#1, or shCdk9#2 measured by flow cytometry at passage 1 after transduction (mean values SEM, n = 3). (H) Phase contrast and AP activity of ESCs in SL or 2iL treated with DMSO or LDC000067 (CDK9i) at the indicated doses. Scale bar, 50 m. (I) As in (H) but shows RT-qPCR result for the indicated genes (mean values SEM, n = 4). (J) As in (F) but ESCs were treated with DMSO or LDC000067. n = 2. A representative experiment is shown. (K) As in (G) but ESCs were treated with DMSO or LDC000067 (mean values SEM, n = 3). (L) Percentage of apoptotic cells in ESCs in 2iL treated with DMSO or LDC000067 (mean values SEM, n = 4). *P < 0.05, **P < 0.01, ***P < 0.001.

BET inhibitors including JQ1 are a promising therapeutic avenue for cancer, but recent reports have described resistance to BET inhibitors through activation of Wnt/-catenin signaling (19, 20). In this pathway, Wnt ligands trigger stabilization and nuclear translocation of -catenin, which then binds to and transactivates T cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors to switch on gene expression (21). We envisaged that -catenin could also confer resistance to BRD4 suppression in ESCs cultured in 2iL, as, similarly to Wnt ligands, CHIR stabilizes -catenin through GSK3 inhibition (3). Moreover, -catenin has been proposed to promote ground-state pluripotency by alleviating the repressor function of TCF3, which associates with pluripotency transcription factors at target loci (2123). Yet, the specific mechanisms are not well understood. Accordingly, Tcf3 or Gsk3 depletion allow expansion of ESCs in serum-free medium with PD alone (3, 21, 23), whereas -catenin is strictly required for expansion in 2iL medium without LIF (21, 22).

We first studied whether PD alone, CHIR alone, or the 2i added to ESCs in SL could rescue the negative effects of 100 nM JQ1 on colony morphology, AP activity, and pluripotency gene expression, which we tested using two ESC lines. PD alone had some rescue effect on Nanog expression but not on the other genes tested. CHIR was more effective in restoring pluripotency characteristics, but only the combined effect of PD and CHIR achieved a complete rescue (Fig. 3, A and B, and fig. S6A). As for cell proliferation, we observed that the moderate rescue effect of adding 2i to ESCs in SL treated with JQ1 was mostly mediated by PD (Fig. 3C).

(A) Phase contrast and AP activity of ESCs cultured in SL with PD, CHIR, or 2i and treated with DMSO or JQ1. Scale bar, 50 m. (B) As in (A) but shows RT-qPCR result for the indicated genes (mean values SEM, n = 3). P value was calculated using two-way ANOVA with Tukeys multiple comparison posttest. (C) Population doublings of ESCs in SL with PD, CHIR, or 2i, and treated with 500 nM JQ1 for 4 days relative to controls treated with DMSO (mean values SEM, n = 4). (D) Phase contrast and AP activity of wild-type (WT) and Gsk3 knockout (KO) ESCs in SL treated with DMSO or JQ1. Scale bar, 50 m. (E) As in (D) but shows RT-qPCR result for the indicated genes (mean values SEM, n = 3). P value was calculated using two-way ANOVA with Sadiks multiple comparison posttest, also for (H), (J), and (L). (F) Heat map showing the fold change of pluripotency (left) and cell cycle genes (right) measured in RNA-seq of wild-type or Gsk3 knockout ESCs in SL treated with DMSO or 100 nM JQ1. (G) Phase contrast and AP activity of wild-type and Tcf3 knockout ESCs in SL treated with DMSO or JQ1. Scale bar, 50 m. (H) As in (G) but shows RT-qPCR result for the indicated genes (mean values SEM, n = 3). (I) Phase contrast and AP activity of wild-type and S33Y -cateninoverexpressing ESCs in SL treated with DMSO or JQ1. Scale bar, 50 m. (J) As in (I) but shows RT-qPCR result for the indicated genes (mean values SEM, n = 3). (K) Phase contrast and AP activity of wild-type and Ctnnb1 knockout ESCs in 2iL treated with DMSO or 200 nM JQ1. Scale bar, 50 m. (L) As in (K) but shows RT-qPCR result for the indicated genes (mean values SEM, n = 3). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

To systematically dissect the role of specific components of the Wnt/-catenin pathway, we treated wild-type ESCs in SL with WNT3A or used several knockout ESC lines lacking either Gsk3 (24), Ctnnb1 (encoding -catenin) (22), or Tcf3 (23). In addition, we used wild-type ESCs overexpressing a mutant form of -catenin (S33Y -catenin) resistant to GSK3-mediated degradation (25). The authentication of Gsk3 and Ctnnb1 knockout ESC lines, and ESCs overexpressing S33Y -catenin, was performed with a -catenin/TCF reporter (fig. S6B), whereas Tcf3 knockout cells were validated by PCR amplification and sequencing (fig. S6C). WNT3A treatment significantly reversed the effects of 100 nM JQ1 on pluripotency characteristics in SL, and Gsk3 knockout achieved a stronger rescue (Fig. 3, D and E, and fig. S6, D and E). The latter was also confirmed by RNA-seq (Fig. 3F and fig. S6F). The stronger effect of Gsk3 knockout compared to WNT3A and CHIR alone is possibly related to the extent and length of GSK3 suppression. Likewise, Tcf3 knockout and S33Y -catenin overexpression induced significant resistance to JQ1 in SL (Fig. 3, G to J). Moreover, Ctnnb1 knockout became sensitive to 200 nM JQ1 in 2iL, but the effect on pluripotency gene expression was not as strong as for wild-type ESCs in SL (Fig. 3, K and L). We also validated the resistance to the CDK9 inhibitor LDC000067 in Gsk3 or Tcf3 knockout ESCs (fig. S6, G and H). Therefore, GSK3 inhibition is the main mediator of the resistance of pluripotency genes to suppression of pause release in 2iL, of which -catenin stabilization is a major component.

To understand how -catenin mediates resistance to suppression of Pol2 pause release at pluripotency loci, we compared -catenin bound sites (table S3) in ChIP-seq (from a study using SL + CHIR) (26) with BRD4 bound sites in 2iL. There was a good genome-wide overlap (Fig. 4A), mostly at distal enhancers but also at promoters (fig. S7A), although the binding of BRD4 was more widespread. Moreover, we noticed that most -catenin/BRD4 cobound genes were not down-regulated by JQ1 in 2iL. We then named -catenin/BRD4 cobound genes that are down-regulated by JQ1 in SL but not 2iL as group 1 genes (Fig. 4B). By contrast, group 2 genes were defined as genes down-regulated by JQ1 in 2iL that are bound by BRD4 but not -catenin. Group 2 included many cell cycle genes, whereas group 1 included many pluripotency regulators (Fig. 4B and table S4). ChIP-qPCR confirmed enhanced -catenin binding at selected group 1 pluripotency loci in 2iL compared to SL, whereas at group 2 cell cyclerelated loci did not change (fig. S7B). ChIP-seq analysis also showed that TCF3 binds to a notable proportion of group 1 genes, whereas most of the group 2 genes were negative (fig. S7C and table S4). These findings suggested that -catenin promotes resistance to Pol2 pause release suppression through cobinding with BRD4/CDK9 at target loci, including pluripotency loci.

(A) Venn diagram showing the overlap between BRD4 bound sites in ESCs in 2iL and -catenin bound sites. (B) Venn diagram showing the overlap between BRD4 bound genes down-regulated by 100 nM JQ1 in ESCs in SL or 2iL and -catenin bound genes. (C) Occupancy plots for genome-wide nuclear run-on sequencing (GRO-seq) signal around the TSS of group 1 and 2 genes in ESCs in SL or 2iL. RPM, reads per million mapped reads. (D) Cumulative plots of GRO-seq signal along the proximal promoter and gene body of group 1 and 2 genes. RPKM, reads per kilobase per million mapped reads. (E) Violin plots showing the corresponding normalized read counts of GRO-seq at the proximal promoter or gene body for group 1 and 2 genes. P value was calculated using Wilcoxon rank sum test, also for all subsequent violin plots and boxplots. (F) As in (E) but shows the TR for group 1 and 2 genes. (G) Genome views from GRO-seq for a pluripotency gene (Nanog) and a cell cycle gene (Stat1) in ESCs in SL or 2iL. (H) ChIP-qPCR for Pol2 Ser5P at the proximal promoter of the indicated pluripotency and cell cycle genes in ESCs in SL or 2iL (mean values SEM, n = 4). (I) As in (H) but shows ChIP-qPCR for Pol2 Ser2P at the gene body (mean values SEM, n = 3). 1 and 2 represent gene body regions 1 and 2, respectively. (J) RT-qPCR for Nanog and Klf2 in ESCs cultured in SL or 2iL treated with 100 or 500 nM THZ1 for the indicated times (mean values SEM, n = 3). (K) RT-qPCR for Nanog and Esrrb in ESCs in 2iL treated with DMSO or 100 nM JQ1 (mean values SEM, n = 3). THZ1 (100 nM) was added for the indicated times before sample collection. *P < 0.05, **P < 0.01, ***P < 0.001.

Next, we sought to elucidate the molecular mechanism underlying the above observations. To rule out the possibility that -catenin compensates for the negative effect of JQ1 on pluripotency genes by enhancing mRNA stability in 2iL (27), we measured a panel of pluripotency mRNAs after actinomycin D treatment, which blocks transcription. Their stability was similar or lower in 2iL compared to SL (fig. S7D). This hinted to -catenin maximizing transcriptional flux at target genes in 2iL as a way to counteract a reduction in Pol2 pause release. So, we turned our attention to potential differences in transcriptional dynamics between ESCs cultured in SL and 2iL. In this regard, a recent Pol2 ChIP-seq study (6) showed a global increase of promoter-proximal signal in 2iL that was not matched in the gene body, concluding that Pol2 pausing is more prevalent in 2iL than in SL. This was attributed to low expression of c-MYC in ESCs in 2iL, as c-MYC induces Pol2 pause release via CDK9 (28). Our reanalysis of this dataset showed a strong increase of Pol2 signal at the proximal promoter in 2iL for both group 1 and group 2 genes (fig. S7, E to G), but this could represent either more Pol2 pausing or more transcriptional initiation. As opposed to Pol2 pausing, more transcriptional initiation implies more gene body elongation if the degree of pausing remains constant and, hence, often associates with increased gene expression. Consistent with the former possibility, the Pol2 signal along the gene body only increased moderately at both groups of genes in 2iL, especially at group 1 (fig. S7, E to G). To define the extent of pausing at these loci more accurately, we used the Pol2 traveling ratio (TR), which compares the ratio in the signal of the proximal promoter and the gene body (12, 28, 29). The TR of both group 1 and group 2 genes was higher in 2iL (fig. S7H), supporting the idea that there is indeed more Pol2 pausing in both groups of genes in 2iL. Yet, it is difficult to reconcile the resistance of group 1 genes to BRD4/CDK9 suppression in 2iL with an increased Pol2 pausing that, in principle, would reduce gene expression. In this regard, we noted several reports describing that, for being a static snapshot, Pol2 ChIP-seq signals cannot effectively distinguish pausing from transcription initiation, nor can they be a measure for effective transcription elongation (29, 30).

We then used genome-wide nuclear run-on sequencing (GRO-seq) (31), which labels nascent RNAs with the synthetic nucleoside 5-bromouridine 5-triphosphate (BrUTP) to accurately map the distribution of transcriptionally engaged Pol2 throughout the genome. Group 1 genes in 2iL displayed enhanced GRO-seq signal not only in the proximal promoter but also in the gene body compared to SL, and the TR did not change significantly (Fig. 4, C to G), together indicating more transcription initiation followed by productive elongation. By contrast, group 2 genes in 2iL showed little difference in the GRO-seq signals in the proximal promoter or the gene body compared to SL, and the TR also remained fairly unchanged (Fig. 4, C to G). The relative pausing level of group 2 genes was higher than that of group 1 genes (Fig. 4F), in agreement with previous reports showing that Pol2 pausing is prevalent at cell cyclerelated genes in both SL and 2iL (29, 32). We also performed ChIP-qPCR for both Pol2 phosphorylated in serine-5 (Ser5P) and Pol2 Ser2P. The former Pol2 modification is mediated by CDK7, a catalytic subunit of the transcription factor H (TFIIH) complex (11), and is considered representative of transcriptional initiation, whereas the latter marks elongating Pol2. Both Pol2 modifications showed a notably increased signal at the Nanog and Esrrb promoter and gene body in 2iL compared with SL (Fig. 4, H and I), but not at two selected cell cyclerelated loci. These results further support the conclusions of the GRO-seq experiment. The GRO-seq of Gsk3 knockout ESCs in SL also showed a pattern consistent with higher transcription initiation and increased gene body elongation at group 1 genes but not group 2 genes compared to the wild-type control (fig. S8, A to D).

We next asked whether the increased transcription initiation at group 1 genes in 2iL may directly contribute to their resistance to Pol2 pause release suppression. Supporting this possibility, higher Pol2 occupancy at the promoter-proximal pause site driven by more transcription initiation can result in greater productive elongation if the function of BRD4/CDK9 is not yet saturated (13). It has also been proposed that increased transcription initiation can nudge paused Pol2 out of the proximal promoter to resume gene body elongation at BRD4 bound genes insensitive to JQ1 (30, 33). To study the relative dependence of pluripotency genes on transcription initiation of ESCs in SL and 2iL, we used the CDK7 inhibitor THZ1 (34). Notably, THZ1 down-regulated pluripotency genes more significantly in 2iL than in SL (Fig. 4J). Moreover, a low dose of THZ1 synergized with 100 nM JQ1 to reduce the expression of pluripotency genes in 2iL (Fig. 4K), supporting the idea that increased initiation compensates for the reduction in pause release mediated by BRD4 inhibition. To study whether the link between Wnt/-catenin signaling, transcription initiation, and JQ1 resistance applies to other contexts, particularly cancer cells, we tested a widely used leukemia cell line, THP1. CHIR induced resistance of THP1 cells to JQ1 (fig. S8E) and also rendered them more sensitive to THZ1 (fig. S8F). Likewise, the combination of THZ1 and JQ1 was more effective than either of the two (fig. S8G). In summary, the recruitment of -catenin to BRD4 bound sites in 2iL changes the mode of transcriptional regulation at target loci including pluripotency loci, which then rely more on transcription initiation for gene body elongation in detriment to Pol2 pause release.

We searched -catenin protein interaction networks looking for partners whose recruitment or reinforcement at group 1 genes in 2iL could explain the above phenomena. In addition to chromatin remodeling complexes (35), we observed two modules corresponding to transcription initiation and elongation (Fig. 5, A and B). Among other -catenin interacting proteins in these modules, we noticed Pol2, TATA-binding proteinassociated factors (TAF5/6/7), cohesin components (SMC1A and SMC3), and, interestingly, BRD4 and CDK9 as well. Pol2 and TAFs are critical for transcription initiation (11), whereas cohesin regulates transcription by forming ring-like structures that allow enhancer-promoter looping (36). We also noticed previous reports describing the interaction of -catenin with mediator (37, 38) and p300 (39) in other cell contexts. Mediator was immediately interesting because it is a well-known partner of BRD4 that controls transcription initiation through both cross-talk with TFIIH and enhancer-promoter looping (40). Immunoprecipitation of -catenin followed by Western blotting confirmed the interaction with mediator (MED1 and MED12), cohesin (SMC1A), and BRD4 in ESCs in 2iL (Fig. 5C). Likewise, ChIP-seq analysis showed genome-wide colocalization of -catenin, MED1, SMC1A, and BRD4 in 2iL at many pluripotency genes belonging to group 1 (Fig. 5D and fig. S9, A and B).

(A) Gene Ontology (GO) analysis of -catenin protein-interactome data based on a previous report (35). GO terms associated with transcriptional regulation and ESC identity are shown (Benjamini-Hochberg corrected P value). (B) Functional network of -catenin interacting proteins related to transcriptional regulation based on STRING protein interaction database (60) as visualized by Cytoscape. -Catenin partners found in both STRING database and the above protein interactome data are highlighted in gray. SMC1A and SMC3 also interact with -catenin but belong to the GO term stem cell population maintenance. (C) Western blotting following immunoprecipitation (IP) of -catenin interacting proteins with nuclear extracts from ESCs in 2iL. Immunoglobulin (IgG) was used as negative control. (D) Genome views of ATAC-seq and H3K27ac, -catenin, MED1, SMC1A, BRD4, and p300 ChIP-seq at Nanog in ESCs cultured as indicated. (E) Occupancy plot (top) and boxplot (bottom) showing the normalized read counts for MED1 ChIP-seq signal in ESCs in SL or SL plus 2i (S2iL) around -catenin bound sites. (F) As in (E) but shows SMC1A ChIP-seq signal. (G) As in (E) but shows BRD4 ChIP-seq signal. (H) ChIP-qPCR for MED1 at -catenin bound sites of the indicated pluripotency genes and cell cycle genes in wild-type and Ctnnb1 knockout ESCs in 2iL (mean values SEM, n = 4). (I) As in (H) but ChIP-qPCR for SMC1A (mean values SEM, n = 5). (J) As in (H) but ChIP-qPCR for BRD4 (mean values SEM, n = 3). (K) RT-qPCR for the indicated genes in Ctnnb1 knockout ESCs in 2iL rescued by knock-in of a wild-type (clone 1) or C-terminal truncated (C) (clone 1) form of -catenin. ESCs were treated with DMSO or 100 nM JQ1 (mean values SEM, n = 3). P value was calculated using two-way ANOVA with Sadiks multiple comparison posttest. (L) As in (C) but uses nuclear extracts from Ctnnb1 knockout ESCs in 2iL rescued by knock-in of a wild-type (clone 1) or a C form (clone 1) of -catenin. Specific bands for IP are marked with red arrows. *P < 0.05, **P < 0.01.

To see whether -catenin is actually promoting the recruitment of these coregulators at target loci, we compared ChIP-seq datasets for MED1, SMC1A, and BRD4 in 2iL and SL. We observed higher levels of the three coregulators at -catenin binding sites in 2iL (Fig. 5, E to G). BRD4 also showed increased signal outside -catenin binding sites but less remarkably, consistent with the idea that the 2i cause a global increase in BRD4 (41). In agreement, ChIP-qPCR for the same three coregulators at -catenin binding sites of pluripotency loci showed reduced levels in Ctnnb1 knockout ESCs in 2iL compared to the wild-type control (Fig. 5, H to J). However, binding of these coregulators at group 2 cell cyclerelated genes remained unchanged. A truncated -catenin form (lacking amino acids 727 to 781) without the C-terminal domain responsible for transcriptional activation of TCF/LEF factors (22) was still competent for inducing resistance to JQ1 in 2iL (Fig. 5K and fig. S9, C and D). This truncated -catenin also retained the ability to interact with coregulators in ESCs in 2iL (Fig. 5L). Overall, our findings support a model in which -catenin strengthens transcriptional flux at pluripotency loci by acting as a scaffold for recruiting coregulators rather than forming a canonical -catenindependent activation complex.

We also investigated chromatin features that could further contribute to maximizing transcriptional flux at group 1 genes in 2iL compared to SL. We focused on histone acetylation and DNA hypomethylation because these epigenetic marks associate with chromatin opening, transcription activation, and reduced Pol2 pausing (42, 43). H3K27 acetylation (H3K27ac) around -catenin binding sites was higher in 2iL than in SL (Figs. 5D and 6A), consistent with the recruitment of histone acetyltransferases (e.g., p300) by -catenin (39). Similarly, we observed an increase in H3K27ac in 2iL when comparing the 2-kb to +2-kb region around the TSS of group 1 genes. By contrast, H3K27ac did not increase at group 2 genes in 2iL compared to SL, and group 2 genes in 2iL had lower H3K27ac than group 1 genes (Fig. 6B). Consistent with the changes in H3K27ac, we noticed a clear increase in open chromatin with an assay for transposase-accessible chromatin sequencing (ATAC-seq) at -catenin binding sites in 2iL compared to SL and more moderately also at group 1 genes, whereas, at group 2 genes, it was slightly reduced in 2iL (Figs. 5D and 6, C and D). Notably, DNA methylation at -catenin binding sites was lower in 2iL than in SL (Fig. 6E). Yet, this effect extended to the entire locus of not only group 1 but also group 2 genes (Fig. 6F), indicating that it is not directly mediated by -catenin. The latter is in agreement with the existence of global DNA hypomethylation in 2iL, which is mostly driven passively through the suppression of UHRF1 protein stability induced by PD (44). In this regard, the limited number of sites actively demethylated by the ten-eleven translocation (TET) enzymes in the conversion of ESCs from SL to 2iL (44) included few -catenin binding sites (fig. S9E). Accordingly, Tet1/2 double and Tet1/2/3 triple knockout ESCs (45) did not show increased sensitivity of pluripotency genes to JQ1 in 2iL compared to the control (fig. S9, F to H). We concluded that permissive chromatin features, some of which are induced by -catenin, likely contribute to strengthening pluripotency gene transcription in 2iL by facilitating the assembly of multiprotein complexes (see schematic in Fig. 7).

(A) Occupancy plot (left) and boxplot (right) showing the normalized read counts for H3K27ac ChIP-seq signal in ESCs in SL or 2iL around -catenin bound sites. (B) Occupancy plot (left and middle) and boxplot (right) showing the normalized read counts for H3K27ac ChIP-seq in ESCs in SL or 2iL around the TSS of group 1 and 2 genes. (C) Occupancy plot (left) and boxplot (right) showing the normalized read counts for ATAC-seq signal in ESCs in SL or 2iL around -catenin bound sites. (D) Occupancy plot (left and middle) and boxplot (right) showing the normalized read counts for ATAC-seq signal in ESCs in SL or 2iL around the TSS of group 1 and 2 genes in ESCs in SL and 2iL. (E) Occupancy plot (left) and boxplot (right) showing the normalized read counts for DNA methylation in ESCs in SL or 2iL around -catenin bound sites. (F) Occupancy plot (left and middle) and boxplot (right) showing the normalized read counts for DNA methylation in ESCs in SL or 2iL around the TSS of group 1 and 2 genes.

(Top) Transcription of pluripotency genes in ESCs in SL requires transcription initiation mediated by recruitment of the Pol2 transcription initiation apparatus, which includes TFIIH, and subsequent pause release mediated by BRD4/CDK9. TCF3 associated with pluripotency transcription factors [including OCT4, SOX2, and NANOG (OSN)] acts as a repressor, presumably by interfering with the proper recruitment of coregulators. (Bottom) In 2iL, -catenin stabilized by GSK3 inhibition is recruited to pluripotency loci. -Catenin facilitates transcription initiation by supplying coregulators including mediator, cohesin, and BRD4, among others, at pluripotent loci. This effect possibly contributes to forming phase-separated condensates resistant to dissociation. The increase in transcription initiation reduces the need for Pol2 pause release mediated by BRD4/CDK9 for productive gene body elongation in 2iL. Higher H3K27ac and DNA hypomethylation renders chromatin genome-wide more accessible in 2iL, potentially facilitating both the recruitment of coregulators and gene body elongation to maximize transcriptional flux at pluripotency genes.

In addition to the recruitment of coregulators and the chromatin changes, other mechanisms may participate in inducing transcriptional resilience at pluripotency loci in 2iL. For example, alternative RNA splicing is a cotranscriptional event that can influence the speed with which Pol2 moves along the gene body (46), and it has also been shown that specific splicing regulators participate in Pol2 pause release (47). Likewise, Gsk3 knockout in ESCs in SL reduces the amount of alternative splicing due to impaired GSK3-mediated phosphorylation of splicing factors (48). We did not observe any notable difference in the number of alternatively spliced genes regulated by GSK3 between group 1 and group 2 genes (fig. S9I). Yet, we noticed that -catenin interacts with multiple splicing regulators including SRSF3 and TRA2B (fig. S9J) (35), both of which also appeared in our screen as differentially required in SL and 2iL (see above Fig. 1A). We validated that Srsf3 and Tra2b knockdown is better tolerated in 2iL compared to SL (fig. S9K). This observation suggests that -catenin helps stabilize splicing regulators at pluripotency genes to render ESCs more resistant to a splicing reduction in 2iL. Although a potential role in modifying the speed of gene body elongation would need to be investigated, these results support the model depicting -catenin as a scaffold that strengthens transcription at pluripotency loci in 2iL.

Mouse ESC pluripotency can be viewed as a continuum of hierarchical interconvertible states on the road to a somatic phenotype. The more nave or closer to inner cell mass characteristics, the more pluripotency is consolidated, but the underlying mechanisms are poorly understood. We have shown here that -catenin stabilized by CHIR selectively reinforces the pluripotency gene network in 2iL by potentiating the recruitment of BRD4, CDK9, mediator, cohesin, p300, and other transcriptional coregulators to pluripotency loci. This selectively heightens transcription initiation at pluripotency loci, enhancing gene body elongation in 2iL and making it morealbeit not completelyindependent of Pol2 pause release by BRD4/CDK9 than in SL. The enhanced transcriptional elongation in 2iL likely explains why expression of multiple pluripotency genes is higher than in SL and potentially also why there is less oscillation in gene expression (an underlying cause of metastability) (7). The removal of TCF3 from pluripotency loci causes a similar transcriptional consequence to -catenin stabilization, conceivably by allowing closer interactions between coregulators and the pluripotency transcription factors or by removing detrimental epigenetic activities [e.g., histone deacetylases (49)]. PD also contributes to inducing resistance to suppression of Pol2 pause release in 2iL possibly by inducing Nanog mRNA and stabilizing NANOG protein (50). The former effect might be caused by preventing extracellular signalregulated kinasemediated phosphorylation and dissociation of coregulators including MED24 from Pol2-containing complexes at the Nanog locus (51). As opposed to pluripotency genes, proliferation genes are not bound by -catenin and, thus, remain very sensitive to suppression of Pol2 pause release in 2iL.

In recent years, it has become evident that phase-separated biomolecular condensates compartmentalize biochemical reactions within cells, including transcription (52). This is caused by multivalent interactions between proteins, many of which have intrinsically disordered regions (IDRs) that confer the physicochemical properties of the condensate. In this regard, it was recently proposed that, thanks to its two disorganized domains at the N-terminal (amino acids 1 to 141) and C-terminal (amino acids 727 to 781) ends, -catenin is attracted to stable chromatin phase-separated condensates formed by mediator and BRD4 to execute its signaling role in ESCs in 2iL (53). Our findings suggest that -catenin might be a priming event for the stabilization of these condensates in ESCs in 2iL by enhancing the cooperative and multivalent interactions between coregulators at pluripotency loci (Fig. 7). This is consistent with our observation that the C-terminal domain of -catenin containing one of its IDRs is not necessary for the resiliance of ESCs in 2iL to JQ1 and the fact that -catenin IDRs are much shorter than those of BRD4 and MED1 (53, 54). The physicochemical forces created within these condensates and the interaction with -catenin could cause a remnant of BRD4, CDK9, and other coregulators to tend to localize to pluripotency loci despite genome-wide depletion induced by shRNAs or chemical inhibitors.

Finley et al. recently reported that BRD4 is dispensable for pluripotency and self-renewal in the ground state (41). A reduced rather than abolished requirement for BRD4 in the early embryo is perhaps easier to understand from a developmental point of view, as it is supported by the observation that Brd4 null mouse embryos cannot maintain the inner cell mass (15, 55). Finley et al. also proposed that a strengthened network of pluripotency transcription factors and the recruitment of TET enzymes partially contribute to the resistance to BRD4 suppression in 2iL. The former mechanism fits well with our observations, as transcription factors can recruit coregulators and enhance transcription initiation (11). Yet, we did not observe any evidence for TET involvement, which may be related to variations among ESC lines or in the culture conditions. Despite the differences, both studies are relevant and highlight the striking similarities in transcriptional adaptation upon network perturbation between ESCs in the ground state and cancer cells. Further mechanistic knowledge will mutually contribute to understand ground-state pluripotency and cancer cell resistance to drugs. For example, ESCs in 2iL may prove to be a useful model to identify either more effective anticancer drugs or synergistic combinations. In this regard, our findings with ESCs in 2iL suggest that treatment of BRD4-addicted cancers with a combination of JQ1 and inhibitors of transcription initiation might be a more robust and applicable anticancer therapy for a general patient base than JQ1 alone.

In the future, it will be important to study whether the molecular interface regulating the interaction between -catenin and transcriptional coregulators can be used to develop specialized anticancer drugs. It will also be interesting to test whether the principles presented here can yield optimized methods for sustaining ground-state pluripotency in vitro in a broad spectrum of mammals.

Human embryonic kidney293T (HEK293T) cells were purchased from the American Type Culture Collection and maintained in Dulbeccos modified Eagles medium (DMEM)/high glucose (Corning, 10-017-CVR) containing 10% fetal bovine serum (FBS; Biowest). THP1 cells were purchased from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) and maintained in RPMI 1640 medium (Thermo Fisher Scientific, C11875500CP) supplemented with 10% FBS (Biowest), GlutaMAX (Gibco, 35050079), penicillin/streptomycin (Hyclone, SV30010), and -mercaptoethanol (Gibco, 2198503). ESCs in SL medium were cultured in DMEM/high glucose containing 15% FBS (Biological Industries; unless otherwise specified), GlutaMAX, penicillin/streptomycin, nonessential amino acids (Gibco, 11140050), sodium pyruvate (Corning, 25-000-CI), -mercaptoethanol, and LIF (1000 U/ml) on mitomycin-Ctreated mouse embryonic fibroblasts (as feeders); they were split onto 0.2% gelatin-precoated plates before each experiment. ESCs in 2iL medium were cultured in a 1:1 mix of DMEM/F12 (Hyclone, SH30023.01) and Neurobasal medium (Gibco, 21103049) with N2 (Gibco, 17502048) and B27 (Gibco, 17504044) supplements, GlutaMAX, penicillin/streptomycin, nonessential amino acids, sodium pyruvate, -mercaptoethanol, LIF (1000 U/ml), 3 M CHIR99021 (StemRD, CHIR-50), and 1 M PD0325901 (StemRD, PD-50) on 0.2% gelatin-precoated plates. SL and 2iL media were changed daily. ESCs cultured in SL medium were cryopreserved in CELLBANKER 2 (Amsbio, 11891). After cell thawing, the same vial was used for culture in SL or 2iL. For the latter, ESCs cultured in SL were adapted to 2iL for three passages before each experiment. ESCs in SL or 2iL were passaged as single cells using 0.05% trypsin (Gibco, 25300054) every 3 days. The other two types of serum for SL medium were purchased from Fisher Scientific and Biowest; both were tested for ESC maintenance beforehand in the Esteban laboratory. Other inhibitors including JQ1 (BPS Bioscience, 27402), LDC000067 (Selleck Chemicals LLC, S7461), THZ1 (MedChemExpress, HY-80013), and actinomycin D (Sigma-Aldrich, A1410) were dissolved in dimethyl sulfoxide and added into the medium at the indicated concentrations. JQ1 and LDC000067 were added for 60 hours unless otherwise specified. E14gt2a (E14) ESCs were provided by I. Samokhvalov (Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, China); they were used for all experiments unless otherwise specified. 129 and OG2 ESCs were provided by J. Liu (Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, China). Tcf3 knockout ESCs (23) were provided by B. Merrill (University of Illinois at Chicago, USA). Gsk3 knockout ESCs, S33Y -cateninoverexpressing ESCs, Ctnnb1 knockout ESCs, Ctnnb1 knockout ESCs rescued by either a wild-type or a C-terminal truncated form of -catenin, Tet1/2 double knockout ESCs, and Tet1/2/3 triple knockout ESCs were previously reported (21, 24, 25, 45) .

For shRNA experiments, ESCs cultured in SL or 2iL medium were infected with lentiviruses generated from HEK293T cells. Samples were extracted 96 hours after infection unless otherwise specified. shRNA inserts were cloned into pLKO.1 lentiviral vectors. All shRNA target sequences and RT-qPCR primers are listed in table S5. RNA samples were isolated using TRIzol reagent (Thermo Fisher Scientific, 15596026). RT-qPCR was performed using the SYBR Premix ExTaq Kit (Takara, RR420A) with an ABI 7500 real-time PCR machine. Data were analyzed in triplicate and normalized on the basis of Actb values. RNA-seq was performed by RiboBio Co. Ltd., China.

Animal experiments were compliant with all relevant ethical regulations regarding animal research and were conducted under the approval of the Animal Care and Use Committee of the Guangzhou Institutes of Biomedicine and Health under license number 2016012. For teratomas, ESCs were trypsinized, and 2 106 cells were injected into the flanks of immunocompromised nude mice. Mice were euthanized when the tumor diameter reached 1.5 cm, and the teratomas were processed for histological analysis. Chimeras were produced by injecting ESCs into blastocysts followed by implantation into a pseudopregnant C57BL/6J mice.

For proliferation assays, 60,000 ESCs were seeded, unless otherwise specified, per well of a six-well plate (three wells per time point). ESCs were counted at the indicated time points with a Bright-Line hemacytometer (Marienfeld). Cell cycle experiments were performed with propidium iodide staining (Beyotime, C1052) followed by flow cytometry analysis. Apoptosis experiments were performed with the Annexin VFITC (fluorescein isothiocyanate) Apoptosis Detection Kit (Vazyme Biotech, A211) followed by flow cytometry analysis. Flow cytometry data were analyzed with FlowJo (v10.4) software. AP activity was detected with the BCIP-NBT Alkaline Phosphatase Color Development Kit (Roche, 11681451001).

Plasmid construction. Dual single guide RNAs (sgRNAs) were designed to target upstream and downstream intron of exon 5, respectively. Two sets of sgRNAs were designed, and the more efficient set was used for the experiments. sgRNAs were cloned into pX330-U6-Chimeric_BB-CBh-hSpCas9 (Addgene, 42230). PKD-EF1-CreER with a puromycin resistance gene was obtained by subcloning pCAG-CreERT2 (Addgene, 14797) into a PKD-EF1 lentiviral backbone plasmid. The left and right homologous arms of the mouse genome and a fragment containing LoxP-exon5-FRT-PGK-Neo-FRT-LoxP were cloned into pMD-19 T donor plasmid (Takara, 6013).

Generation of Brd4fl/fl clones. ESCs cultured in 2iL medium were transduced with the donor and PX330-CAS9-sgRNA plasmids using Lipofectamine 3000 (Invitrogen, L3000015). G418 (Merk, 108321-42-2) was added 24 hours after transduction for selection. After selection, the remaining cells were seeded into a 96-well plate for genotyping. To obtain Brd4fl/fl clones, the remaining cells were again transfected with pCAG-FlpeGFP plasmid (Addgene, 13788) and the green fluorescent protein (GFP)positive cells were sorted out to remove the selective marker Neo that was already integrated. Cells were then transduced with the donor and PX330-CAS9-sgRNA plasmids for a second round. After selection with G418, all the remaining cells were seeded again into a 96-well plate for genotyping. For Brd4fl/fl clones, the left-LoxP-exon5containing fragment and right-LoxPcontaining fragment were amplified for Sanger sequencing to make sure that the sequence and position of the LoxPs and exon 5 were correctly modified.

Generation of Brd4 fl/ and Brd4/ clones. Brd4fl/fl clones were transduced with the PKD-EF1-CreER plasmid and selected with puromycin (InvivoGen, ant-pr-1) for 2 days. The expression level of CreER was tested by RT-qPCR. Cells were seeded into a 96-well plate, and 4-hydroxytamoxifen was added to induce deletion of the floxed alleles. Genotyping was performed to obtain Brd4 fl/ and Brd4/ clones. Brd4 fl/ clones were transduced with pCAG-CreGFP plasmid (Addgene, 13776), and GFP-positive cells were sorted 72 hours later. The sorted GFP-positive cells were seeded into a 96-well plate for genotyping to get Brd4/ clones. All primers are listed in table S5.

Ten million cells were cross-linked in freshly prepared formaldehyde solution (1% final concentration for 10 min at room temperature) and then quenched with 125 mM glycine (for 5 min at room temperature). Fixed cells were washed with cold phosphate-buffered saline (PBS), harvested, flash-frozen in liquid nitrogen, and stored at 80C for further use. For -catenin, Pol2 Ser5P, and Pol2 Ser2P ChIP-qPCR, immunoprecipitation was performed as reported by Ward et al. (56). For MED1, SMC1A, and BRD4 ChIP-qPCR, immunoprecipitation was performed as reported by Finley et al. (41). After elution of antibody-bound complexes from the beads, cross-linking was reversed by overnight incubation at 65C. Samples were diluted in TE (Tris-EDTA) buffer and then treated with ribonuclease A (Sigma-Aldrich, R6513) for 1 hour at 37C, followed by incubation with proteinase K (Thermo Fisher Scientific, 25530049) for 2 hours at 55C. DNA was purified using the QIAquick PCR Purification Kit (Qiagen, 28106). Antibodies used for ChIP-qPCR were immunoglobulin G (Abcam, ab172730), anti-catenin (Abcam, ab32572), antiPol2 Ser5P (Abcam, ab5131), antiPol2 Ser2P (Abcam, ab5095), anti-MED1 (Bethyl, A300-793), anti-SMC1A (Bethyl, A300-055), and anti-BRD4 (Bethyl, A301-985A). Primers for ChIP-qPCR are listed in table S5.

GRO-seq was performed as previously described (57). Briefly, nuclei from 107 ESCs were extracted and run-on-transcribed with BrUTP (Sigma-Aldrich, B7166) and other nucleoside 5-triphosphates at 30C for 5 min. Nascent RNA was enriched by agarose-coated anti-BrUTP (Santa Cruz Biotechnology, sc-32323). Poly(A) tail was added to the nascent RNA by poly(A) polymerase (New England Biolabs, M0276S) to synthesize complementary DNA with oligo(dT) primers. GRO-seq libraries were amplified by PCR for 10 cycles and separated with 10% tris-borate EDTA polyacrylamide gels. Bands ranging from 160 to 300 base pairs (bp) were cut and purified by isopropanol precipitation. Sequencing of GRO-seq libraries was performed by Berry Genomics Co. Ltd., China.

Cells (107) were lysed in 250 l of TNE lysis buffer [50 mM tris-HCl (pH 7.5), 250 mM NaCl, 0.5% NP-40, and 1 mM EDTA] containing protease inhibitor cocktail (Roche, 04693132001) on ice for 15 min. Lysates were homogenized by a 0.4-mm syringe needle and centrifuged at 13,000g for 15 min at 4C. Supernatants were diluted with TNEG buffer [50 mM tris-HCl (pH 7.5), 50 mM NaCl, 0.5% NP-40, 20% glycerol, and 1 mM EDTA] and then incubated with the relevant antibodies overnight at 4C. The next day, 30 l of prewashed Protein A/Gconjugated beads (Thermo Fisher Scientific, 10001D and 10003D) was added and incubated for 3 hours at 4C. Beads were then washed three times with wash buffer 1 [20 mM tris-HCl (pH 7.4), 125 mM NaCl, and 0.1% NP-40] and two times with wash buffer 2 (1 PBS with 0.02% NP-40) for 5 min under rotation at 4C (for each wash). Last, the proteins were eluted with 60 l of SDS loading buffer and boiled for Western blotting. The following primary antibodies were used for immunoprecipitation or Western blotting: anti-BRD4 (Bethyl, A301-985A), anti-tubulin (Sigma-Aldrich, T5201), anti-catenin (Abcam, ab32572), anti-MED1 (Bethyl, A300-793), anti-MED12 (Bethyl, A300-774A), and anti-SMC1A (Bethyl, A300-055).

Cells (50,000) were washed once with cold PBS and resuspended in 50 l of lysis buffer [10 mM tris-HCl (pH 7.4) 10 mM NaCl, 3 mM MgCl2, and 0.1% (v/v) IGEPAL CA-630]. The suspension was then centrifuged at 500g for 10 min at 4C, followed by addition of 50 l of transposition reaction mix of the TruePrep DNA Library Prep Kit (Vazyme Biotech, TD502). Samples were then incubated at 37C for 30 min. Transposition reactions were cleaned up using the MinElute PCR Purification Kit (Qiagen, 28004). ATAC-seq libraries were subjected to five cycles for preamplification and amplified by PCR for an appropriate number of cycles. The amplified libraries were purified using the QIAquick PCR Purification Kit (Qiagen, 28104). Library concentration was measured using the VAHTSTM Library Quantification Kit (Vazyme Biotech, NQ101). Libraries were sequenced by Berry Genomics Co. Ltd., China.

For RNA-seq gene expression quantification, data were first aligned with STAR (v2.5.2) and quantified according to GENCODE vM15 in an RSEM-based pipeline (58). Differentially expressed genes were determined by DESeq2 (v1.18.1) and were defined as absolute fold change of >2 and q value of <0.1. Functional annotation was further performed by ClusterProfiler (v3.6.0) (59). For ChIP-seq and ATAC-seq, data were first aligned to the mm10 mouse genome assembly using Bowtie2 (v2.2.5) with the settings --very-sensitive. Low-quality mapped reads were removed using Samtools with the settings -q 30. Duplicated reads were collapsed using Picard (v1.9.0). For ChIP-seq, binding sites were called using MACS2 (v2.1.0) with the settings --keep-dup 1 -q 0.01. Peaks were annotated to all genes within 2 kb and the single closest gene within 20 kb, and duplicate genes were removed. Peaks were considered as overlapping if they intersect with each other. For GRO-seq, adaptors were first trimmed with fastp (v0.20.0), and only read 1 was kept for further analysis. PCR duplicates were collapsed using FASTX-Toolkit. A 20-bp polyA sequence and an 8-bp random sequence were trimmed from 3 end. Clean data were then aligned to the mm10 mouse genome assembly using Bowtie2. For quantification of Pol2 ChIP-seq and GRO-seq signals, the proximal promoter was considered as the 100- to +300-bp region around the annotated TSS, and the gene body was considered as the +300-bp to +2-kb region downstream of the annotated TSS. Reads were first normalized as reads per million mapped reads or reads per kilobase per million mapped reads using deepTools (v 3.3.1) and further assigned to the corresponding regions, while the top 1% of the values were trimmed. For whole-genome bisulfite sequencing analysis, data were aligned to the mm10 mouse genome assembly using BSMAP with the settings -v 0.1 -g 1 -p 8 -R -u and further assigned to corresponding regions. Occupancy plots were generated by deepTools. Cumulative plots, violin plots, and boxplots were generated by ggplot2 (v2.2.1); the black central line of boxplots is the median, the boxes indicate the upper and lower quartiles, and the whiskers indicate the 1.5 interquartile range.

ESCs were seeded 12 hours before transfection on gelatin-precoated 24-well plates at a density of 30,000 cells per well. TOPflash or FOPflash report plasmids (Millipore, 17-285) and Renilla luciferase plasmids were transduced using Lipofectamine 3000. Twenty-four hours after transfection, luciferase activity was measured using a Dual-Luciferase Reporter Assay System (Promega, E1910).

CCK-8 Cell Counting Kit (Vazyme Biotech, A311-02) was used to evaluate the cell viability of THP1 cells. THP1 cells (8,000 per well) were seeded in a 96-well plate. For measurements, 10 l of CCK-8 solution was added to each well, and the plates were incubated for 1 to 4 hours at 37C before the absorbance was measured at 450 nm using an Epoch 2 microplate spectrophotometer from BioTek.

Data of bar charts are represented as mean SEM. The P value was calculated using the unpaired two-tailed Students t test or two-way analysis of variance (ANOVA). The number of replicates for each experiment is indicated in the figure legends. For violin plots and boxplots, the P value was calculated using Wilcoxon rank sum test.

Acknowledgments: We thank all members of the Esteban laboratory for their comments. We also thank M. Oren (Weizmann Institute of Science, Israel) for technical advice and J. T. Lis (Cornell University, USA) and X. Fu (University of California, USA) for helpful comments on this manuscript. We also thank the technical support from the Guangzhou Branch of the Supercomputing Center of Chinese Academy of Science and the Experimental Animal Center of Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences. Funding: This work was supported by the National Key Research and Development Program of China (2016YFA0100102, 2016YFA0100701, 2016YFA0100300, and 2018YFA0106903), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDA16030502), the National Natural Science Foundation of China (31671537, 31571524, 31501192, 31430049, 31850410463, 31970619, 31950410553, and 31900617), the Guangdong Province Science and Technology Program (2014A030312001, 2015A030308007, 2016B030229007, 2016A050503037, and 2017B050506007), the Guangzhou Science and Technology Program (201807010066), the Innovative Team Program of Guangzhou Regenerative Medicine and Health Guangdong Laboratory (2018GZR110103001), and the Science and Technology Planning Project of Guangdong Province, China (2017B030314056). J.H.H. was funded by Pascal and Ilana Mantoux, Helen and Martin Kimmel Institute for Stem Cell Research, Flight Attendant Medical Research Council (FAMRI), European Research Council (ERC-CoG), and an Israel-China Israel Science Foundation (ISF) grant. C.W. was supported by a Zhujiang Overseas Young Talents Postdoctoral Fellowship. S.K. was supported by a Chinese Academy of Sciences Presidents International Fellowship. M.M.A., D.P.I., and M.T. were supported by the Chinese Academy of SciencesThird World Academy of Sciences (TWAS) Presidents PhD Fellowship. A.S. was supported by the Deutsche Forschungsgemeinschaft (REBIRTH and SFB738). Author contributions: M.A.E., M.Z., and Y. Lai conceived the idea and designed the experiments. M.Z. conducted most of the experiments and Y. Lai performed most of the bioinformatics study. M.A.E., M.Z., and Y. Lai analyzed the data. V.K. and L.C. contributed critically to the experiments. P.G., X.G., Jianguo Zhou, Y.X., Z.Y., L.L., A.J., W.L., M.M.A., G.M., N.L., X.F., Y. Lv., M.J., M.T., S.K., H.L., X.X., H.Z., Y.H., L.W., S.C., I.A.B., Z.L., D.W., T.Z., C.W., M.H., D.P.I., Y. Li, Jiajian Zhou, J.Y., Y.F., K.A., U.D.V., F.G., A.P.H., and G.V. contributed to the experiments and/or the analyses. X.B., G.W., A.S., H.W., H.S., B.Q., A.P.H., B.W.D., C.H., M.P.C., Y.Q., G.-L.X., R.C., and G.V. provided relevant advice, essential materials, and/or infrastructural support. M.A.E. supervised the study and provided most of the financial support. J.H.H. contributed to the supervision and also provided financial support. M.A.E. wrote the manuscript with help from M.Z. and Y. Lai. M.A.E. approved the final version of the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors. RNA-seq, GRO-seq, and ATAC-seq data have been deposited in the Gene Expression Omnibus database under the accession number GSE123692. Published datasets used in this study are listed in table S6.

Read this article:
-Catenin safeguards the ground state of mousepluripotency by strengthening the robustness of the transcriptional apparatus - Science Advances

COVID-19 Impact on Cell Therapy Industry 2020: Global Market Size, Share, Emerging Trends, Business Growth Applications, SWOT Analysis by Top Key…

TheGlobal Cell Therapy Marketwas estimated to be valued at USD XX million in 2018 and is projected to reach USD XX million by 2026, at a CAGR of XX% during 2019 to 2026. Growing aging patient population, the rise in cell therapy transplantations globally, and rising disease awareness drive the growth of the market.

Get Sample Copy of this Report @https://www.orianresearch.com/request-sample/1281174

What you can expect from our report: Total Addressable Market [ Present Market Size forecasted to 2026 with CAGR ] Regional level split [North America, Europe, Asia Pacific, South America, Middle East & Africa] Country wise Market Size Split [Important countries with major market share] Market Size Breakdown by Product/ Service Types [ ] Market Size by Application/Industry verticals/ End Users [ ] Market Share and Revenue/Sales of 10-15 Leading Players in the Market Production Capacity of Leading Players whenever applicable Market Trends Emerging Technologies/products/start-ups, PESTEL Analysis, SWOT Analysis, Porters Five Forces, etc. Pricing Trend Analysis Average Pricing across regions Brandwise Ranking of Major Market Players globally

Cell therapyinvolves the administration of somatic cell preparations for the treatment of diseases or traumatic damages. The objective of this study is to provide long term treatment through a single injection of therapeutic cells.

However, stringent regulatory policies may restrain growth of the market in the forecast period.

The global Cell Therapy Market is primarily segmented based on different type, technique, cell source, technology, end users and region.

On the basis of type, the market is split into: * Allogenic Therapies * Autologous Therapies

On the basis of technique, the market is split into: * Stem Cell Therapy * Cell Vaccine * Adoptive Cell Transfer (ACT) * Fibroblast Cell Therapy * Chondrocyte Cell Therapy * Other Technique

On the basis of cell source, the market is split into: * Bone Marrow * Adipose Tissue * Umbilical Cord Blood-Derived Cells

On the basis of technology, the market is split into: * Viral Vector Technology * Cell Immortalization Technology * Genome Editing Technology

On the basis of end user, the market is split into: * Hospitals & Clinics * Regenerative Medicine Centers * Other End Users

Inquire more or share a question if any before the purchase on this report @https://www.orianresearch.com/enquiry-before-buying/1281174

Moreover, the market is classified based on regions and countries as follows: * North America- U.S., Canada * Europe- U.K., France, Germany, Italy and Rest of Europe * Asia-Pacific- China, Japan, India and Rest of Asia Pacific * South America- Brazil, Mexico and Rest of South America * Middle East & Africa- South Africa, Saudi Arabia and Rest of Middle East & Africa

Key Market Players: The key players profiled in the market include: * Kolon TissueGene, Inc. * JCR Pharmaceuticals Co. Ltd. * Osiris Therapeutics, Inc. * Stemedica Cell Technologies, Inc. * Fibrocell Science, Inc. * Vericel Corporation * Pharmicell Co., Ltd * Anterogen.CO.,LTD. * Baxter Healthcare Corporation. * Arteriocyte Medical Systems

These enterprises are focusing on growth strategies, such as new product launches, expansions, acquisitions, and agreements & partnerships to expand their operations across the globe.

Key Benefits of the Report: * Global, regional, country, type, technique, cell source, technology, end users market size and their forecast from 2018-2026 * Identification and detailed analysis on key market dynamics, such as, drivers, restraints, opportunities, and challenges influencing growth of the market * Detailed analysis on industry outlook with market specific PESTLE, and supply chain to better understand the market and build expansion strategies * Identification of key market players and comprehensively analyze their market share and core competencies, detailed financial positions, key product, and unique selling points * Analysis on key players strategic initiatives and competitive developments, such as joint ventures, mergers, and new product launches in the market * Expert interviews and their insights on market shift, current and future outlook, and factors impacting vendors short term and long term strategies * Detailed insights on emerging regions, type, technique, cell source, technology and end users with qualitative and quantitative information and facts

Target Audience: * Cell Therapy Manufactures * Traders, Importers, and Exporters * Raw Material Suppliers and Distributors * Research and Consulting Firms * Government and Research Organizations * Associations and Industry Bodies

Research Methodology: The market is derived through extensive use of secondary, primary, in-house research follows by expert validation and third party perspective, such as, analyst reports of investment banks. The secondary research is the primary base of our study wherein we conducted extensive data mining, referring to verified data products, such as, white papers, government and regulatory published articles, technical journals, trade magazines, and paid data products.

For forecasting, regional demand & supply factors, recent investments, market dynamics including technical growth scenario, consumer behavior, and end use trends and dynamics, and production capacity were taken into consideration. Different weightages have been assigned to these parameters and quantified their market impacts using the weighted average analysis to derive the market growth rate.

The market estimates and forecasts have been verified through exhaustive primary research with the Key Industry Participants (KIPs), which typically include: * Manufacturers * Suppliers * Distributors * Government Body & Associations * Research Institutes

Get Direct Copy of this Report @https://www.orianresearch.com/checkout/1281174

Table of Content 1. Introduction 2. Research Methodology 3. Executive Summary 4. Global Cell Therapy Market Overview 4.1. Market Segmentation & Scope 4.2. Market Trends 4.2.1. Drivers 4.2.2. Restraints 4.2.3. Opportunities 4.2.4. Supply Chain Analysis 4.3. Global Cell Therapy Market Porters Five Forces Analysis 4.4. Global Cell Therapy Market PESTEL Analysis 5. Global Cell Therapy Market, by Type 5.1. Global Cell Therapy Market, Size and Forecast, 2015-2026 5.2. Global Cell Therapy Market, by Allogenic Therapies, 2015-2026 5.2.1. Key driving factors, trends and opportunities 5.2.2. Market size and forecast, 2015-2026 5.3. Global Cell Therapy Market, by Autologous Therapies, 2015-2026 5.3.1. Key driving factors, trends and opportunities 5.3.2. Market size and forecast, 2015-2026 6. Global Cell Therapy Market, by Technique 6.1. Global Cell Therapy Market, Size and Forecast, 2015-2026 6.2. Global Cell Therapy Market, by Stem Cell Therapy, 2015-2026 6.2.1. Key driving factors, trends and opportunities 6.2.2. Market size and forecast, 2015-2026 6.3. Global Cell Therapy Market, by Cell Vaccine, 2015-2026 6.3.1. Key driving factors, trends and opportunities 6.3.2. Market size and forecast, 2015-2026

..Continued

Customization Service of the Report:

Orian Research provides customisation of reports as per your need. This report can be personalised to meet your requirements. Get in touch with our sales team, who will guarantee you to get a report that suits your necessities.

About Us:

Orian Research is one of the most comprehensive collections of market intelligence reports on the World Wide Web. Our reports repository boasts of over 500000+ industry and country research reports from over 100 top publishers. We continuously update our repository so as to provide our clients easy access to the worlds most complete and current database of expert insights on global industries, companies, and products. We also specialize in custom research in situations where our syndicate research offerings do not meet the specific requirements of our esteemed clients.

Contact Us:

Ruwin Mendez

Vice President Global Sales & Partner Relations

Orian Research Consultants

US: +1 (832) 380-8827 | UK: +44 0161-818-8027

Email:[emailprotected]

Website: http://www.orianresearch.com/

Read the original post:
COVID-19 Impact on Cell Therapy Industry 2020: Global Market Size, Share, Emerging Trends, Business Growth Applications, SWOT Analysis by Top Key...

Somatic Cells: Meaning, Characteristics, Types and Examples

Somatic cells account for all the cells of the body except reproductive cells. Other than gametes, stem cells and germs cells, all the cells of a multicellular organism are known as somatic cells.

Diploid somatic cells undergo mitosis and are responsible for growth, repair and regeneration.

Somatic Cells Meaning

Somatic terms originate from the word Soma, which means body. They make up the entire organism other than cells, which have a reproductive function or are undifferentiated, e.g. stem cells.

Somatic Cells Characteristics

Somatic Cells Types and Examples

There are numerous types of somatic cells. In our body, there are 220 types of somatic cells. Many cells are differentiated to perform various specific functions.

Some of the specialised somatic cells are:

Explore all the important topics aligned with the updated NEET syllabus, only at BYJUS. Check NEET Important topics and Preparation Tips for all the important concepts and related topics.

Read more from the original source:
Somatic Cells: Meaning, Characteristics, Types and Examples

Takeda and the New York Academy of Sciences Announce 2020 Innovators in Science Award Winners – BusinessGhana

The 2020 award celebrates outstanding research in rare diseases Takeda Pharmaceutical Company Limited (Takeda) (TSE:4502/NYSE:TAK) and the New York Academy of Sciences announced today the Winners of the third annual Innovators in Science Award for their excellence in and commitment to innovative science that has significantly advanced the field of rare disease research.

Each Winner receives a prize of US $200,000.

This press release features multimedia.

View the full release here: https://www.

businesswire.

com/news/home/20200708005039/en/ The 2020 Winner of the Senior Scientist Award is Adrian R.

Krainer, Ph.

D.

, St.

Giles Foundation Professor at Cold Spring Harbor Laboratory.

Prof.

Krainer is recognized for his outstanding research on the mechanisms and control of RNA splicing, a step in the normal process by which genetic information in DNA is converted into proteins.

Prof.

Krainer studies splicing defects in patients with spinal muscular atrophy (SMA), a devastating, inherited pediatric neuromuscular disorder caused by loss of motor neurons, resulting in progressive muscle atrophy and eventually, death.

Prof.

Krainers work culminated notably in the development of the first drug to be approved by global regulatory bodies that can delay and even prevent the onset of an inherited neurodegenerative disorder.

Collectively, rare diseases affect millions of families worldwide, who urgently need and deserve our help.

Im extremely honored to receive this recognition for research that my lab and our collaborators carried out to develop the first approved medicine for SMA, said Prof.

Krainer.

As basic researchers, we are driven by curiosity and get to experience the thrill of discovery; but when the fruits of our research can actually improve patients lives, everything else pales in comparison.

The 2020 Winner of the Early-Career Scientist Award is Jeong Ho Lee, M.

D.

, Ph.

D, Associate Professor, Korea Advanced Institute of Science and Technology (KAIST).

Prof.

Lee is recognized for his research investigating genetic mutations in stem cells in the brain that result in rare developmental brain disorders.

He was the first to identify the causes of intractable epilepsies and has identified the genes responsible for several developmental brain disorders, including focal cortical dysplasias, Joubert syndromea disorder characterized by an underdevelopment of the brainstemand hemimegalencephaly, which is the abnormal enlargement of one side of the brain.

Prof.

Lee also is the Director of the National Creative Research Initiative Center for Brain Somatic Mutations, and Co-founder and Chief Technology Officer of SoVarGen, a biopharmaceutical company aiming to discover novel therapeutics and diagnosis for intractable central nervous system (CNS) diseases caused by low-level somatic mutation.

It is a great honor to be recognized by a jury of such globally respected scientists whom I greatly admire, said Prof.

Lee.

More importantly, this award validates research into brain somatic mutations as an important area of exploration to help patients suffering from devastating and untreatable neurological disorders.

The 2020 Winners will be honored at the virtual Innovators in Science Award Ceremony and Symposium in October 2020.

This event provides an opportunity to engage with leading researchers, clinicians and prominent industry stakeholders from around the world about the latest breakthroughs in the scientific understanding and clinical treatment of genetic, nervous system, metabolic, autoimmune and cardiovascular rare diseases.

At Takeda, patients are our North Star and those with rare diseases are often underserved when it comes to the discovery and development of transformative medicines, said Andrew Plump, M.

D.

, Ph.

D.

, President, Research & Development at Takeda.

Insights from the ground-breaking research of scientists like Prof.

Krainer and Prof.

Lee can lead to pioneering approaches and the development of novel medicines that have the potential to change patients lives.

Thats why we are proud to join with the New York Academy of Sciences to broadly share and champion their workand hopefully propel this promising science forward.

Connecting science with the world to help address some of societys most pressing challenges is central to our mission, said Nicholas Dirks, Ph.

D.

, President and CEO, the New York Academy of Sciences.

In this third year of the Innovators in Science Award we are privileged to recognize two scientific leaders working to unlock the power of the genome to bring innovations that address the urgent needs of patients worldwide affected by rare diseases.

About the Innovators in Science Award The Innovators in Science Award grants two prizes of US $200,000 each year: one to an Early-Career Scientist and the other to a well-established Senior Scientist who have distinguished themselves for the creative thinking and impact of their research.

The Innovators in Science Award is a limited submission competition in which research universities, academic institutions, government or non-profit institutions, or equivalent from around the globe with a well-established record of scientific excellence are invited to nominate their most promising Early-Career Scientists and their most outstanding Senior Scientists working in one of four selected therapeutic fields of neuroscience, gastroenterology, oncology, and regenerative medicine.

Prize Winners are determined by a panel of judges, independently selected by the New York Academy of Sciences, with expertise in these disciplines.

The New York Academy of Sciences administers the Award in partnership with Takeda.

For more information please visit the Innovators in Science Award website.

About Takeda Pharmaceutical Company Limited Takeda Pharmaceutical Company Limited (TSE:4502/NYSE:TAK) is a global, values-based, R&D-driven biopharmaceutical leader headquartered in Japan, committed to bringing Better Health and a Brighter Future to patients by translating science into highly-innovative medicines.

Takeda focuses its R&D efforts on four therapeutic areas: Oncology, Rare Diseases, Neuroscience, and Gastroenterology (GI).

We also make targeted R&D investments in Plasma-Derived Therapies and Vaccines.

We are focusing on developing highly innovative medicines that contribute to making a difference in people's lives by advancing the frontier of new treatment options and leveraging our enhanced collaborative R&D engine and capabilities to create a robust, modality-diverse pipeline.

Our employees are committed to improving quality of life for patients and to working with our partners in health care in approximately 80 countries.

For more information, visit https://www.

takeda.

com.

About the New York Academy of Sciences The New York Academy of Sciences is an independent, not-for-profit organization that since 1817 has been committed to advancing science, technology, and society worldwide.

With more than 20,000 members in 100 countries around the world, the Academy is creating a global community of science for the benefit of humanity.

The Academy's core mission is to advance scientific knowledge, positively impact the major global challenges of society with science-based solutions and increase the number of scientifically informed individuals in society at large.

Please visit us online at www.

nyas.

org.

.

Read more:
Takeda and the New York Academy of Sciences Announce 2020 Innovators in Science Award Winners - BusinessGhana