Report: Austin Aries DDP Trying To Get AEW To Buy Into Stem Cell Treatment Venture – Fightful

Austin Aries was backstage at AEW Dynamite in Atlanta, GA last night and according to a new report, he was pitching a stem cell treatment venture.

According to Cassidy Haynes of Bodyslam.net, Aries and Diamond Dallas Page were at AEW Dynamite trying to get AEW to buy into a stem cell treatment venture that they are part of.

Wrestlers have taken to stem cell treatment in past few years with Rey Mysterio, Brian Cage, John Morrison, Rob Van Dam, and more undergoing stem cell treatment to help prolong their careers and deal with past injuries. Aries has mentioned BioXcellerator stem cell therapy on his social media in the past.

Recently, Aries has been hanging out with Page and taken to DDP Yoga. Page has a DDP Yoga studio based in Georgia and is a regular at AEW events due to his relationship with Cody Rhodes. Page has also been featured on AEW television in past months, including wrestling a match on the Bash at the Beach (Jan. 15) episode of AEW Dynamite.

Aries last wrestled for Major League Wrestling and is currently a free agent.

If you missed anything from last night's AEW Dynamite, you can check out Fightful's full report on the show by clicking here.

Read more:
Report: Austin Aries DDP Trying To Get AEW To Buy Into Stem Cell Treatment Venture - Fightful

On the Trail of Cancer Stem Cells – Technology Networks

Two research teams from the Max Delbrck Center for Molecular Medicine and their collaborators have produced a detailed cell atlas of an entire salivary gland tumor in a mouse model, mapping individual cells throughout the tumor and its surrounding tissue. The "single cell" approach, recently described inNature Communications, has provided key insights about cellular composition changes through the earliest stages of cancer development.

A solid tumor is not, as many might assume, a lump of cells that are all the same. Rather it is mix of many different cell types, including a variety of stromal and immune cells besides the actual tumor cells.

"Conventional methods in molecular biology often consider a sample as a whole, which fails to recognize the complexity within it," said Dr. Samantha Praktiknjo, senior scientist and first author from MDC's Systems Biology of Gene Regulatory Elements Lab headed by Professor Nikolaus Rajewsky at the Berlin Institute for Medical Systems Biology (BIMSB). Developing a detailed understanding of the different cells within a tumor and how they interact could help identify more effective treatment strategies.Strength in numbers

The team used single-cell RNA sequencing technologies developed in theRajewsky laband novel epitope profiling to produce the cell atlas, and identified the cells that were specific to the tumor by leveraging the reproducibility and the large sample size of their data.

The latter was possible by using a mouse model, developed in MDC's Signal Transduction in Development and Cancer Lab headed by Professor Walter Birchmeier, which harbors designed mutations that induce a salivary gland squamous cell carcinoma. This system provides a consistent supply of genetically similar tumors to sequence from the earliest stages of development, which is nearly impossible with human patients.

"In a patient, the tumor is already developed and you cannot go back and rewind time and look at how it started," said Dr. Benedikt Obermayer, a co-first author now at the Berlin Institute of Health (BIH). "Here, we have a model that is so controlled, we can watch it happen." And Dr. Qionghua Zhu, the third first author and a former postdoc at theBirchmeier Lab, added: "To fight cancer effectively, we need to find the driver mutations. This method gives us clues about the evolution trajectories of a tumor."

Sequencing technologies have advanced so that it is now possible to quickly and affordably sequence the RNA inside single cells, one at a time, as well as the proteins on the surfaces of cells in the tissues. While other methods grind up the tissue and identify what genes and molecules are present in the mix, the single cell approach precisely identifies how many of each type of cell is present, and which genes and molecules are associated with which cell.

For this study, the researchers sequenced more than 26,000 individual salivary gland cells from mice with tumors and healthy mice. They used computational models to analyze the huge amount of data and identify each individual cell and sort them into groups - such as stromal cells, immune cells, saliva producing cells, cancer cells - based on the hundreds of genes expressed and molecules present.A surprise

The single cell approach revealed something that surprised the researchers: "When I saw the data, I thought, where is the tumor?" Obermayer said. The population of cancer stem cells in the tumor was extremely small - less than one percent of all profiled cells in the tissue. Due to their low abundance, investigation of these cells still heavily depends on assumptions about surface markers and is often performed in cell culture-based systems. Here, the authors were able to identify the cancer stem cells directly from the solid tumor samples with their single cell approach.

Furthermore, the team was able to predict the progression of the different cell types through different stages of tumor development. Their model suggests that the cancer stem cells emerge from cancerous basal cells, then develop into another subtype before ultimately becoming a population of cells similar to luminal cells, a cell type present in normal, healthy salivary glands.

This progression supports the idea that when something goes awry in the basal cells of this solid tumor model, they are triggered to turn into cancer stem cells, which can then become a different type of cell. "What I found fascinating was clearly seeing the order of signals and events, transitioning from the progenitor to the progeny populations of the cancer stem cells," Praktiknjo said.Next steps

Further research is required to verify that individual cells are transforming through these stages, and explore the cellular and molecular interactions driving tumor growth. The team anticipates the approach they've demonstrated here can be applied to other cancer types as well.

"To me the main conceptual insight is that we can apply ideas from single-cell based developmental biology to reconstruct molecular progression of tumorigenesis ," said Professor Nikolaus Rajewsky, who heads MDC's Systems Biology of Gene Regulatory Elements Lab and is the scientific director of the BIMSB.

Reference:Praktiknjo, et al. (2020) Tracing tumorigenesis in a solid tumor model at single-cell resolution, Nature Communications, DOI: 10.1038/s41467-020-14777-0.

This article has been republished from the following materials. Note: material may have been edited for length and content. For further information, please contact the cited source.

View post:
On the Trail of Cancer Stem Cells - Technology Networks

Dutch startup Neuroplast raises 4M for its stem cell-based technology to treat patients with Spinal Cord Injury – Silicon Canals

Neuroplast is a company based in Maastricht (the Netherlands) developing autologous stem cell therapies for patients suffering from neurodegenerative diseases such as spinal cord injury (SCI), amyotrophic lateral sclerosis (ALS) and traumatic brain injury.

Recently, the company has raised 4 million from Dutch-based Brightlands Venture Partners and LIOF and from an existing shareholder and informal investor Lumana Invest BV.

CEO Johannes de Munter said:

The financing and support of the investors will enable us to perform multicenter clinical trials in the Netherlands, Denmark, Germany, and Spain and bring the product to market.

This Dutch startup will use the fund to perform a phase II/III clinical trial with the aim of obtaining conditional market approval for the treatment of patients suffering from Spinal Cord Injury.

Founded by physician Hans de Munter and neurologist Erik Wolters in 2014, Neuroplast has expanded with Juliette van den Dolder, who was appointed as COO and management team member.

In the case of SCI, isolating, manufacturing, and reinserting patients own cells, very promising preclinical outcomes have resulted in an Orphan Drug Designation from European regulatory authorities, allowing a fast-track procedure for the clinical trials. These trials are expected to start in March 2020.

Marcel Kloosterman Director at Brightlands Venture Partners:

Neuroplast combines breakthrough science with a solid management team. In a sizable market characterised by major unmet need, successful treatment of (accident caused) paralysed patients would make life so much easier for them and their families while lowering the burden and costs for the society.

Yearly, 24,500 people in Europe and the USA are diagnosed with Spinal Cord Injury, usually caused by accident. Its worth mentioning that for Europe and the US, the medical cost associated with Spinal Cord Injury is over 13 bn per year.

CEO Johannes de Munter adds:

Neuroplast is becoming an ATMP player in the region and wants to contribute to our beautiful eco-system.

Main image credits:Neuroplast

Stay tuned toSilicon Canalsfor more European technology news

Go here to read the rest:
Dutch startup Neuroplast raises 4M for its stem cell-based technology to treat patients with Spinal Cord Injury - Silicon Canals

Animal Stem Cell Therapy Market 2020-2026: Product Types, by Applications, By Market Trends, Market Reserach Report – Keep Reading

Our latest research report entitle Global Animal Stem Cell Therapy Market provides comprehensive and deep insights into the market dynamics and growth of Global Animal Stem Cell Therapy Industry. Latest information on market risks, industry chain structure, Animal Stem Cell Therapy cost structure and opportunities are offered in this report. The entire industry is fragmented based on geographical regions, a wide range of applications and Global Animal Stem Cell Therapy Market types. The past, present and forecast market information will lead to investment feasibility by studying the crucial Global Animal Stem Cell Therapy Industry growth factors.

Get FREE Sample Report Copy @ https://www.globalmarketers.biz/report/life-sciences/2018-global-animal-stem-cell-therapy-industry-research-report/118224 #request_sample

Global Animal Stem Cell Therapy Market Analysis By Major Players:

VETSTEM BIOPHARMAMediVet BiologicJ-ARMCelavetMagellan Stem CellsU.S. Stem CellCells Power JapanANIMAL CELL THERAPIESAnimal Care StemCell Therapy SciencesVetCell TherapeuticsAnimacelAratana Therapeutics

Global Animal Stem Cell Therapy Market Analysis By Geographical Zones:

Europe Market (Germany, France, Italy, Russia and UK)

North America Market (Canada, USA and Mexico)

Latin America Market (Middle and Africa).

Animal Stem Cell Therapy Market in Middle East and Africa (Saudi Arabia, UAE, Egypt, Nigeria and South Africa)

Asia-Pacific Market (South-east Asia, China, India, Korea and Japan).

Global Animal Stem Cell Therapy Market news, plans & policies, market drivers, analysis of upstream raw material suppliers and downstream buyers of Animal Stem Cell Therapy is carried out in this report. Global Animal Stem Cell Therapy Industry forecast study enlists the market value (in USD) and volume forecast for each region, product type, and applications.

Types Of Global Animal Stem Cell Therapy Market:

DogsHorsesOthers

Applications Of Global Animal Stem Cell Therapy Market:

Veterinary HospitalsResearch Organizations

Inquiry Here For Detail Report @ https://www.globalmarketers.biz/report/life-sciences/2018-global-animal-stem-cell-therapy-industry-research-report/118224 #inquiry_before_buying

To Provide A Clear Global Animal Stem Cell Therapy Market Structure The Report Is Divided Into 12 Chapters As Follows:

Explore Full Report With Detailed TOC Here @ https://www.globalmarketers.biz/report/life-sciences/2018-global-animal-stem-cell-therapy-industry-research-report/118224 #table_of_contents

Major points from Table of Contents for Global Animal Stem Cell Therapy Industry 2020 Market Research Report include:

1 Market Overview

2 Global Animal Stem Cell Therapy Market Competition by Manufacturers

3 Global Animal Stem Cell Therapy Capacity, Production, Revenue (Value) by Region (2020-2026)

4 Global Animal Stem Cell Therapy Industry Supply (Production), Consumption, Export, Import by Region (2020-2026)

5 Global Animal Stem Cell Therapy Production, Revenue (Value), Price Trend by Type

6 Global Animal Stem Cell Therapy Market Analysis by Application

7 Global Animal Stem Cell Therapy Industry Manufacturers Profiles/Analysis

8. Animal Stem Cell Therapy Manufacturing Cost Analysis

9 Industrial Chain, Sourcing Strategy and Downstream Buyers

10 Marketing Strategy Analysis, Distributors/Traders

11 Market Effect Factors Analysis

12 Global Animal Stem Cell Therapy Market Forecast (2020-2026)

13 Research Findings and Conclusion

14 Appendix

Explore Full Report With Detailed TOC Here @ https://www.globalmarketers.biz/report/life-sciences/2018-global-animal-stem-cell-therapy-industry-research-report/118224 #table_of_contents

Read more:
Animal Stem Cell Therapy Market 2020-2026: Product Types, by Applications, By Market Trends, Market Reserach Report - Keep Reading

Global demand of Stem Cell Therapy Market will boom in coming years – Nyse Nasdaq Live

Stem Cell Therapy Market Presented and Promoted by Report CMI pulls historical, traditional and forecast market essential value of Stem Cell Therapy industry by 2026, report highlights opportunities, regional markets, emerging growth factors, market c hospitality, forecasts and competitors Joined the market share. The primary purpose of the Stem Cell Therapy market report is to provide a proper and strategic analysis of the Stem Cell Therapy industry.

The market structure covers the value chain, player category, product range, the presence of key players across the product, and the end user segment of the market. The report also provides a snapshot of the major competition, market trend forecasts and forecasts over the next five years, projected growth rates and key factors affecting driving and growth market data and analysis are derived from a combination of primary and secondary sources.

Look at the Free Sample Copy Report: https://www.coherentmarketinsights.com/insight/request-sample/2848

The study included research on a variety of factors affecting the industry, including government policies, market conditions, competitive environments, historical data, current market trends, technological innovations, technological advances in future technologies and related industries, and market risks.Opportunities, market barriers and challenges.

Best Merchant Analysis is one of the key components and is very useful for each player to understand the focused scene in the market. The main major companies in the Stem Cell Therapy market report are: Osiris Therapeutics, Inc. Novartis AG, GlaxoSmithKline Plc., MEDIPOST Co., Ltd., Anterogen Co., Ltd. Pharmicell Co., Ltd. Holostem Terapie Avanzate S.r.l. JCR Pharmaceuticals Co., Ltd. NuVasive, Inc. RTI Surgical, Inc., and Fibrocell Science, Inc.

The Stem Cell Therapy Market Report is a reliable source for market research that will exponentially accelerate your business. It also provides new challenges SWOT testing, speculative feasibility studies and venture return surveys.

Market Driver

Market Challenges

Market Trends

Purchase Copy of This Business Report: https://www.coherentmarketinsights.com/insight/buy-now/2848

Key Questions on the Stem Cell Therapy Market 20202026:

What will the market size be in 2026 and what will the growth rate be?

What are the main market trends?

What are the main factors driving this market?

What are the challenges to market growth?

Who are the main major suppliers in this market?

What are the market opportunities, market risks and market overview threats faced by major suppliers?

What are the Strengths and Weaknesses of the Major Suppliers?

After all, the Stem Cell Therapy Market Report provides conclusions that include research findings, market size estimates, market share, changes in consumer demand / customer preferences, and data sources. These factors improve the overall business.

Download the Free PDF Here: https://www.coherentmarketinsights.com/insight/request-pdf/2848

About us:

Coherent Market Insights is a prominent market research and consulting firm that offers action-ready syndicate reports, custom market analysis, consulting services and competitive analysis, with a variety of recommendations related to emerging market trends, technologies and potential absolute dollar opportunities.

Contact Us:Name: Mr.ShahPhone: US +12067016702 / UK +4402081334027Email: [emailprotected]Visit our Blog: https://hospitalhealthcareblog.wordpress.com/

See the original post:
Global demand of Stem Cell Therapy Market will boom in coming years - Nyse Nasdaq Live

Combination Enfortumab Vedotin + Pembrolizumab Granted Breakthrough Therapy in Bladder Cancer – OncoZine

The U.S. Food and Drug Administration (FDA) has granted Breakthrough Therapy designation to enfortumab vedotin-ejfv (Padcev; Astellas Pharma and Seattle Genetics) in combination with Mercks (known as MSD outside the United States and Canada) anti-PD-1 therapy pembrolizumab (Keytruda) for the treatment of patients with unresectable locally advanced or metastatic urothelial cancer who are unable to receive cisplatin-based chemotherapy in the first-line setting.

It is estimated that approximately 81,000 people in the U.S. will be diagnosed with bladder cancer in 2020. [1] Urothelial cancer accounts for 90% of all bladder cancers and can also be found in the renal pelvis, ureter, and urethra. [2] Globally, approximately 549,000 people were diagnosed with bladder cancer in 2018, and there were approximately 200,000 deaths worldwide. [3]

The recommended first-line treatment for patients with advanced urothelial cancer is cisplatin-based chemotherapy. For patients who are unable to receive cisplatin, such as people with kidney impairment, a carboplatin-based regimen is recommended. However, fewer than half of patients respond to carboplatin-based regimens and outcomes are typically poorer compared to cisplatin-based regimens. [4]

Conditionally approvedEnfortumab vedotin-ejfv, a first-in-class antibody-drug conjugate (ADC) that is directed against Nectin-4, a protein located on the surface of cells and highly expressed in bladder cancer, was conditionally approved by the FDA in December 2019 based on the Accelerated Approval Program. [5][6]

Antibody-drug Conjugates or ADCs are highly targeted biopharmaceutical drugs that combine monoclonal antibodies specific to surface antigens present on particular tumor cells with highly potent anti-cancer agents linked via a chemical linker.

With seven approved drugs on the market, ADCs have become a powerful class of therapeutic agents in oncology and hematology.

Continued approval for enfortumab vedotin-ejfv in combination with pembrolizumab for the treatment of patients with advanced or metastatic urothelial cancer may be contingent upon verification and description of clinical benefit in confirmatory trials. [5]

The drug is indicated for the treatment of adult patients with locally advanced or metastatic urothelial cancer who have previously received a programmed death receptor-1 (PD-1) or programmed death-ligand 1 (PD-L1) inhibitor and a platinum-containing chemotherapy before (neoadjuvant) or after (adjuvant) surgery or in a locally advanced or metastatic setting.

Nonclinical data suggest the anticancer activity of enfortumab vedotin is due to its binding to Nectin-4 expressing cells followed by the internalization and release of the anti-tumor agent monomethyl auristatin E (MMAE) into the cell, which result in the cell not reproducing (cell cycle arrest) and in programmed cell death (apoptosis). [5]

Breakthrough therapyThe Breakthrough Therapy process is designed to expedite the development and review of drugs that are intended to treat a serious or life-threatening condition. The designation is based upon preliminary clinical evidence indicating that the drug may demonstrate substantial improvement over available therapies on one or more clinically significant endpoints. In the case of enfortumab vedotin, the designation was based on the initial results from Phase Ib/II EV-103 Clinical Trial.

The FDAs Breakthrough Therapy designation reflects the encouraging preliminary evidence for the combination of enfortumab vedotin and pembrolizumab in previously untreated advanced urothelial cancer to benefit patients who are in need of effective treatment options, said Andrew Krivoshik, M.D., Ph.D., Senior Vice President, and Oncology Therapeutic Area Head, Astellas.

We look forward to continuing our work with the FDA as we progress our clinical development program as quickly as possible.

This is an important step in our investigation of enfortumab vedotin in combination with pembrolizumab as first-line therapy for patients with advanced urothelial cancer who are unable to receive cisplatin-based chemotherapy, said Roger Dansey, M.D., Chief Medical Officer, Seattle Genetics.

Based on encouraging early clinical activity, we recently initiated a phase III trial of this platinum-free combination and look forward to potentially addressing an unmet need for patients.

Clinical trialThe Breakthrough Therapy designation was granted based on results from the dose-escalation cohort and expansion cohort A of the Phase Ib/II trial, EV-103 (NCT03288545), evaluating patients with locally advanced or metastatic urothelial cancer who are unable to receive cisplatin-based chemotherapy-treated in the first-line setting with enfortumab vedotin-ejfv in combination with pembrolizumab.

The initial results from the trial were presented at the European Society of Medical Oncology (ESMO) 2019 Congress, and updated findings at the 2020 Genitourinary Cancers Symposium.

EV-103 is an ongoing, multi-cohort, open-label, multicenter phase Ib/II trial of PADCEV alone or in combination, evaluating the safety, tolerability, and efficacy in muscle-invasive, locally advanced and first- and second-line metastatic urothelial cancer.

Adverse eventsSerious adverse reactions occurred in 46% of patients treated with enfortumab vedotin-ejfv. The most common serious adverse reactions (3%) were urinary tract infection (6%), cellulitis (5%), febrile neutropenia (4%), diarrhea (4%), sepsis (3%), acute kidney injury (3%), dyspnea (3%), and rash (3%). Fatal adverse reactions occurred in 3.2% of patients, including acute respiratory failure, aspiration pneumonia, cardiac disorder, and sepsis (each 0.8%).

Discontinuing treatmentAdverse reactions leading to discontinuation occurred in 16% of patients; the most common adverse reaction leading to discontinuation was peripheral neuropathy (6%). Adverse reactions leading to dose interruption occurred in 64% of patients; the most common adverse reactions leading to dose interruption were peripheral neuropathy (18%), rash (9%) and fatigue (6%). Adverse reactions leading to dose reduction occurred in 34% of patients; the most common adverse reactions leading to dose reduction were peripheral neuropathy (12%), rash (6%) and fatigue (4%).

The most common adverse reactions (20%) were fatigue (56%), peripheral neuropathy (56%), decreased appetite (52%), rash (52%), alopecia (50%), nausea (45%), dysgeusia (42%), diarrhea (42%), dry eye (40%), pruritus (26%) and dry skin (26%). The most common Grade 3 adverse reactions (5%) were rash (13%), diarrhea (6%) and fatigue (6%).

Specific recommendations

HyperglycemiaHyperglycemia occurred in patients treated with enfortumab vedotin-ejfv, including death and diabetic ketoacidosis (DKA), in patients with and without pre-existing diabetes mellitus. The incidence of Grade 3-4 hyperglycemia increased consistently in patients with higher body mass index and in patients with higher baseline A1C. In one clinical trial, 8% of patients developed Grade 3-4 hyperglycemia. Patients with baseline hemoglobin A1C 8% were excluded.

Physicians are recommended to closely monitor blood glucose levels in patients with, or at risk for, diabetes mellitus or hyperglycemia and, if blood glucose is elevated (>250 mg/dL), withhold the drug.

Peripheral neuropathyPeripheral neuropathy (PN), predominantly sensory, occurred in 49% of the 310 patients treated with enfortumab vedotin-ejf in clinical trials. Two percent (2%) of patients experienced Grade 3 reactions. In one clinical trial, peripheral neuropathy occurred in patients treated with enfortumab vedotin-ejf with or without preexisting peripheral neuropathy.

The median time to onset of Grade 2 was 3.8 months (range: 0.6 to 9.2). Neuropathy led to treatment discontinuation in 6% of patients. At the time of their last evaluation, 19% had complete resolution, and 26% had partial improvement.

Physicians should:

Occular disordersOcular disorders occurred in 46% of the 310 patients treated with enfortumab vedotin-ejf. The majority of these events involved the cornea and included keratitis, blurred vision, limbal stem cell deficiency and other events associated with dry eyes. Dry eye symptoms occurred in 36% of patients, and blurred vision occurred in 14% of patients, during treatment with enfortumab vedotin-ejf.

The median time to onset to symptomatic ocular disorder was 1.9 months (range: 0.3 to 6.2).

Physicians should monitor patients for ocular disorders and consider:

Skin reactionsSkin reactions occurred in 54% of the 310 patients treated with enfortumab vedotin-ejf in clinical trials. Twenty-six percent (26%) of patients had a maculopapular rash and 30% had pruritus. Grade 3-4 skin reactions occurred in 10% of patients and included symmetrical drug-related intertriginous and flexural exanthema (SDRIFE), bullous dermatitis, exfoliative dermatitis, and palmar-plantar erythrodysesthesia. In one clinical trial, the median time to onset of severe skin reactions was 0.8 months (range: 0.2 to 5.3).

Of the patients who experienced rash, 65% had complete resolution and 22% had partial improvement.

Physicians should monitor patients for skin reactions, and consider:

Infusion site extravasationSkin and soft tissue reactions secondary to extravasation have been observed after the administration of enfortumab vedotin-ejf. Of the 310 patients, 1.3% of patients experienced skin and soft tissue reactions. Reactions may be delayed.

Erythema, swelling, increased temperature, and pain worsened until 2-7 days after extravasation and resolved within 1-4 weeks of peak. One percent (1%) of patients developed extravasation reactions with secondary cellulitis, bullae, or exfoliation.

Physicians should ensure adequate venous access prior to starting enfortumab vedotin-ejf and monitor for possible extravasation during administration. If extravasation occurs, stop the infusion and monitor for adverse reactions.

Embryo-fetal toxicityEnfortumab vedotin-ejf can cause fetal harm when administered to a pregnant woman.

Physicians should advise patients of the potential risk to the fetus and advise female patients of reproductive potential to use effective contraception during enfortumab vedotin-ejf treatment and for 2 months after the last dose. At the same time, they should advise male patients with female partners of reproductive potential to use effective contraception during treatment with enfortumab vedotin-ejf and for 4 months after the last dose.

Clinical trialA Study of Enfortumab Vedotin Alone or With Other Therapies for Treatment of Urothelial Cancer (EV-103) NCT03288545

References[1] American Cancer Society. Cancer Facts & Figures 2020. Online. Last accessed on January 23, 2020.[2] American Society of Clinical Oncology. Bladder cancer: introduction (10-2017). Online. Last accessed on January 23, 2020.[3] International Agency for Research on Cancer. Cancer Tomorrow: Bladder. Online. Last accessed on January 23, 2020.[4] National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology: Bladder Cancer. Version 4; July 10, 2019. Online. Last accessed on January 23, 2020.[5] Enfortumab vedotin-ejfv (Padcev; Astellas Pharma [package insert]. Northbrook, IL)[6] Challita-Eid P, Satpayev D, Yang P, et al. Enfortumab Vedotin Antibody-Drug Conjugate Targeting Nectin-4 Is a Highly Potent Therapeutic Agent in Multiple Preclinical Cancer Models. Cancer Res 2016;76(10):3003-13.

A version of this article was first published in ADC Review | Journal of Antibody-drug Conjugates.

Go here to see the original:
Combination Enfortumab Vedotin + Pembrolizumab Granted Breakthrough Therapy in Bladder Cancer - OncoZine

Vascular endotheliumtargeted Sirt7 gene therapy rejuvenates blood vessels and extends life span in a Hutchinson-Gilford progeria model – Science…

Abstract

Vascular dysfunction is a typical characteristic of aging, but its contributing roles to systemic aging and the therapeutic potential are lacking experimental evidence. Here, we generated a knock-in mouse model with the causative Hutchinson-Gilford progeria syndrome (HGPS) LmnaG609G mutation, called progerin. The Lmnaf/f;TC mice with progerin expression induced by Tie2-Cre exhibit defective microvasculature and neovascularization, accelerated aging, and shortened life span. Single-cell transcriptomic analysis of murine lung endothelial cells revealed a substantial up-regulation of inflammatory response. Molecularly, progerin interacts and destabilizes deacylase Sirt7; ectopic expression of Sirt7 alleviates the inflammatory response caused by progerin in endothelial cells. Vascular endotheliumtargeted Sirt7 gene therapy, driven by an ICAM2 promoter, improves neovascularization, ameliorates aging features, and extends life span in Lmnaf/f;TC mice. These data support endothelial dysfunction as a primary trigger of systemic aging and highlight gene therapy as a potential strategy for the clinical treatment of HGPS and age-related vascular dysfunction.

Aging represents the largest risk factor for many age-related diseases, as exemplified by cardiovascular diseases (CVDs) (1). The blood vessel consists of the tunica intima [composed of endothelial cells (ECs)], the tunica media [composed of vascular smooth muscle cells (VSMCs)], and the tunica adventitia (consisting of connective tissue) (2). The endothelium separates the vessel wall from blood flow and has an irreplaceable role in regulating vascular tone and homeostasis. Age-related functional decline in ECs and VSMCs is a main cause of CVDs (3). ECs secrete various vasodilators and vasoconstrictors that act on VSMCs and induce blood vessel contraction and relaxation (4). For instance, nitric oxide (NO) is synthesized from l-arginine by endothelial NO synthase (eNOS) and then released on VSMCs to induce blood vessel relaxation (5). When ECs become senescent or dysfunctional, vasoconstrictive, procoagulative, and proinflammatory cytokines are released; this effect reduces NO bioavailability and, in turn, increases vascular intimal permeability and EC migration (6). Despite advances in the understanding of mechanisms of endothelial dysfunction, it is unclear whether it directly triggers organismal aging.

Accumulating evidences suggest that the mechanisms underlying physiological aging are similar to those governing Hutchinson-Gilford progeria syndrome (HGPS)a premature aging syndrome in which affected patients typically succumb to CVDs (7). HGPS is predominantly caused by an a.c. 1824 C>T, p. G608G mutation in LMNA gene, which activates an alternate splicing event and generates a 50amino acid truncated form of Lamin A, referred to as progerin (8). The murine LmnaG609G, which is equivalent to LMNAG608G in humans, causes aging phenotypes resembling HGPS (9). It has been shown that progerin targets SMCs and causes blood vessel calcification and atherosclerosis (10, 11). Recent work by two groups showed that SMC-specific progerin knock-in (KI) mice are healthy and have a normal life span but suffer from blood vessel calcification, atherosclerosis, and shortened life span when crossed to Apoe/ mice (12, 13). In contrast to SMCs, the contributing roles of the vascular endothelium (VE) to systemic/organismal aging are still elusive. To address these issues, we generated a conditional progerin (LmnaG609G) KI model, i.e., Lmnaf/f mice. In combination with E2A-Cre and Tie2-Cre mice, in which the expression of Cre is ubiquitous including germ cells (14) or driven by the endothelial-specific Tie2 promoter (15), we aimed to investigate the roles of VE dysfunction to systemic aging and the targeting potential for the clinical treatment of HGPS.

To study the mechanism of VE aging, we generated a mouse model of conditional progerin KI, in which the LmnaG609G mutation, equivalent to HGPS LMNAG608G, was flanked with loxP sites, i.e., Lmnaf/f mice (fig. S1A). The Lmnaf/f mice were crossed to E2A-Cre mice, in which the Cre recombinase is ubiquitously expressed including germ cells, to generate LmnaG609G/G609G and LmnaG609G/+ mice. Progerin was ubiquitously expressed in LmnaG609G/G609G and LmnaG609G/+ mice, which recapitulated many progeroid features found in HGPS, including growth retardation and shortened life span (fig. S1, B to D).

To understand primary alterations in the VE, we isolated CD31+ murine lung ECs (MLECs) (16) from three pairs of LmnaG609G/G609G (G609G) and Lmnaf/f (Flox) mice by fluorescence-activated cell sorting (FACS) (Fig. 1A) and performed 10 Genomics single-cell RNA sequencing. We recovered 6004 cells (4137 from G609G and 1867 from Flox mice) and used the k-means clustering algorithm to cluster the cells into four groups (Fig. 1B). As expected, one group exhibited high Cd31, Cd34, and Cdh5 expression and thus largely represented MLECs. The other three groups, copurified with CD31+ MLECs by FACS, showed relatively lower Cd31 expression at the mRNA level (>10-fold lower than MLECs) but high Cd45 expression (fig. S2). Further analysis revealed that these clusters most likely contained B lymphocytes (B-like) with high Cd22, Cd81, and Ly6d expression; T lymphocytes (T-like) with high Cd3d, Cd3e, and Cd28 expression; and macrophages (M-like) with high Cd14, Cd68, and Cd282 expression (Fig. 1C). Most of the marker gene expression levels were comparable between G609G and Flox mice, except for Cd34 and Icam1, which were significantly elevated in G609G ECs, and Cd14 and Vcam1, which were increased in G609G M-like cells (Fig. 1D). Of note, Icam1 and Vcam1 are among the most conserved markers of endothelial senescence and atherosclerosis (17). Thus, we established an Lmnaf/f conditional progerin KI mouse model and revealed a unique EC population for mechanistic study.

(A) Purity analysis of sorted CD31+ MLECs by FACS. SSC, side scatter; FSC, forward scatter; PE, phycoerythrin. (B) t-Distributed stochastic neighbor embedding (t-SNE) projection of CD31+ cells revealed four clusters: ECs (green), B lymphocytes (B-like; orange), T lymphocytes (T-like; blue), and macrophages (M-like; red). (C) Marker gene expression in the four clusters: ECs (Cd31, Cd34, and Cdh5), B-like (Ly6d, Cd22, and Cd81), T-like (Cd3d, Cd3e, and Cd28), and M-like (Cd14, Cd68, and Cd282). (D) Heatmap showing marker gene expression levels in LmnaG609G/G609G (G609G) and Lmnaf/f (Flox) mice.

Of the four clusters of CD31+ MLECs, ECs and M-like cells showed high levels of p21Cip1/Waf1 (fig. S2A), a typical senescence marker (18). This finding suggests that these cells are the main target of progerin in the context of aging. A previous study reported that M-specific progerin, achieved by crossing Lmnaf/+ to Lyz-Cre mice, caused minimal aging phenotypes (12), implicating that M might have only a minor role in organismal aging. We thus focused on ECs for further analysis. We recovered 899 and 445 ECs from E2A and Flox mice, respectively (Fig. 2A). Genes with >1.5-fold change in expression between these mice were chosen for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. We observed a significant enrichment in the pathways that regulate chemotaxis, immune responses in malaria and Chagas diseases, inflammatory bowel disease, and rheumatoid arthritis and pathways essential for cardiac function (Fig. 2, B to D). To confirm this observation and to exclude paracrine effects from other cell types, we overexpressed progerin in human umbilical vein ECs (HUVECs) and analyzed representative genes by quantitative polymerase chain reaction (PCR). Most of the examined genes, e.g., IL6, IL8, IL15, CXCL1, IL1, etc., were significantly up-regulated upon ectopic progerin overexpression (Fig. 2E). Together, these data suggest that progerin causes an inflammatory response in VE, which might lead to systemic aging.

(A) t-SNE projection of LmnaG609G/G609G (G609G; green) and Lmnaf/f (Flox; orange) CD31+ MLECs according to transcriptomic data. (B to D) GO and KEGG pathway enrichment of differentially expressed genes between G609G and Flox cells. LmnaG609G/G609G MLECs show enrichment in genes that regulate the inflammatory response (C) and genes related to heart dysfunction (D). FC, fold change; FDR, false discovery rate. (E) Quantitative PCR analysis of altered genes observed in (C) and (D) in HUVECs with ectopic expression of progerin or wild-type LMNA. Data represent means SEM. *P < 0.05, **P < 0.01, and ***P < 0.001 (Students t test).

To test whether the VE dysfunction has essential roles in systemic aging, we crossed Lmnaf/f mice to a Tie2-Cre line to generate Lmnaf/f;TC mice, in which the expression of Cre recombinase is driven by the promoter/enhancer of endothelial-specific Tie2 gene (15). Single-cell transcriptome analysis confirmed that Tie2 was mainly detected in ECs (fig. S2B). Consistently, progerin was observed in the VE of Lmnaf/f;TC, but not in that of Lmnaf/f control mice or other tissues (fig. S3). VE-specific progerin induced intima-media thickening in Lmnaf/f;TC mice, in a similar manner to total KI mice, i.e., LmnaG609G/G609G mice (Fig. 3, A and B). We performed functional analysis of the VE based on acetylcholine (ACh)regulated vasodilation. ACh-induced thoracic aorta relaxation was significantly compromised in Lmnaf/f;TC mice (Fig. 3C). Similar defects were observed in LmnaG609G/G609G and LmnaG609G/+ mice (Fig. 3D and fig. S4), where progerin was expressed in both ECs and SMCs (12). To gain more evidence supporting VE-specific dysfunction, we examined thoracic aorta relaxation induced by sodium nitroprusside (SNP), which is an SMC-dependent vasodilator. Little difference was observed in thoracic aorta vasodilation in LmnaG609G/G609G and LmnaG609G/+ compared to Lmnaf/f control mice (Fig. 3E and fig. S4), supporting the notion that the VE dysfunction is a key contributor of vasodilation defects in progeria mice. As NO is the most potent vasodilator (19), we examined eNOS levels in the thoracic aorta of Lmnaf/f;TC and Lmnaf/f control mice. As expected, the level of eNOS was significantly reduced in Lmnaf/f;TC mice compared to Lmnaf/f control mice (Fig. 3F). Thus, the data confer a VE-specific dysfunction in progeria mice.

(A and B) Hematoxylin and eosin staining of thoracic aorta sections from (A) Lmnaf/f;TC and (B) LmnaG609G/G609G and Lmnaf/f control mice showing intima-media thickening. Scale bar, 20 m. (C) ACh-induced thoracic aorta vasodilation in Lmnaf/f;TC and Lmnaf/f control mice. **P < 0.01. 5-HT, 5-hydroxytryptamine. (D) ACh-induced thoracic aorta vasodilation in LmnaG609G/G609G and control mice. **P < 0.01. (E) SNP-induced thoracic aorta vasodilation in LmnaG609G/G609G and control mice. (F) eNOS level in thoracic aorta sections from Lmnaf/f;TC and control mice. Scale bar, 20 m. (G) Immunofluorescence staining (left) and quantification (right) of CD31+ gastrocnemius muscle in Lmnaf/f;TC and Lmnaf/f mice. Scale bar, 50 m. DAPI, 4,6-diamidino-2-phenylindole. (H) CD31 immunofluorescence staining in Lmnaf/f;TC and Lmnaf/f liver. Scale bar, 50 m. (I) Representative microcirculation images (left) and quantification of blood flow recovery (right) following hindlimb ischemia in Lmnaf/f;TC and Lmnaf/f mice. (J) Representative transverse sections and quantification of CD31+ gastrocnemius muscle 14 days after femoral artery ligation. Scale bar, 50 m. All data represent means SEM. P values were calculated by Students t test. Photo credits: Shimin Sun, School of Life Sciences, Shandong University of Technology; Medical Research Center, Shenzhen University (A, B, F, H, and J); Weifeng Qin, Medical Research Center, Shenzhen University (G and I).

The reduced capillary density and neovascularization capacity are both characteristics of endothelial dysfunction (1). We examined the microvasculature in various tissues of Lmnaf/f;TC mice by immunofluorescence staining. We observed a significant loss in CD31+ ECs in Lmnaf/f;TC mice compared to controls (Fig. 3, G and H). We further examined ischemia-induced neovascularization ability in Lmnaf/f;TC mice following femoral artery ligation. Limb perfusion after ischemia was significantly blunted in Lmnaf/f;TC mice compared to controls (Fig. 3I). Histological analysis confirmed that the defect in blood flow recovery in Lmnaf/f;TC mice was a reflection of an impaired ability to form new blood vessels in the ischemic region (Fig. 3J). Together, Lmnaf/f;TC mice are characterized by a loss of ECs, a reduced capillary density, and defective neovascularization capacity.

The single-cell transcriptome implicates heart dysfunction in LmnaG609G/G609G mice (Fig. 2). A correlation with gene alterations associated with atherosclerosis and osteoporosis was obvious in LmnaG609G/G609G ECs (the Online Mendelian Inheritance in Man; https://omim.org) (fig. S5). We thus reasoned that endothelial-specific dysfunction might be enough to trigger systemic aging. Notably, atherosclerosis was prominent in Lmnaf/f;TC mice (aorta atheromatous plaque observed in all nine examined mice; Fig. 4A), as well as severe fibrosis in the arteries and hearts (Fig. 4, B and C); both are typical features of aging. Moreover, the heart/body weight ratio was significantly increased in Lmnaf/f;TC compared to Lmnaf/f control mice (Fig. 4D), indicating dilated cardiomyopathy (20). Echocardiography confirmed that heart rate, cardiac output, left ventricular ejection fraction, and fractional shortening were significantly reduced in 7- to 8-month-old Lmnaf/f;TC compared to Lmnaf/f control mice. The running endurance was largely compromised in Lmnaf/f;TC mice (Fig. 4E), which is likely a reflection of amyotrophy. Moreover, the microcomputed tomography (CT) identified a decrease in trabecular bone volume/tissue volume, trabecular thickness, and trabecular number but an increase in trabecular separation in Lmnaf/f;TC mice (Fig. 4F), indicative of osteoporosis, which is an important hallmark of systemic aging (21). The VE-specific dysfunction not only accelerated aging in various tissues/organs but also shortened the median life span of Lmnaf/f;TC mice (24 weeks) to a similar extent to LmnaG609G/G609G mice (21 weeks) (Fig. 4G). LmnaG609G/G609G mice suffered from body weight loss roughly from 8 weeks of age, while Lmnaf/f;TC mice only showed a slight drop in body weight (Fig. 4H), suggesting that body weight loss itself is a less likely primary causal factor to progeria compared to endothelial dysfunction. Together, these results implicate that endothelial dysfunction, at least in progeria, acts as a causal factor of systemic aging.

(A to C) Masson trichrome staining showing an atheromatous plaque in the aorta (A), SMC loss (B), and cardiac fibrosis (C) in Lmnaf/f;TC mice. Scale bar, 20 m. (D) Heart weight and echocardiographic parameters, including heart rate, cardiac output, left ventricular (LV) ejection fraction (LVEF), and left ventricular ejection shortening (LVFS). *P < 0.05, Lmnaf/f;TC versus Lmnaf/f mice. (E) Decreased running endurance in Lmnaf/f;TC mice. ***P < 0.001. (F) Micro-CT analysis showing a decrease in trabecular bone volume/tissue volume (BV/TV), trabecular number, and trabecular thickness and an increase in trabecular separation in Lmnaf/f;TC mice. *P < 0.05, Lmnaf/f;TC versus Lmnaf/f mice. (G) Life span of LmnaG609G/G609G, LmnaG609G/+, Lmnaf/f;TC, and Lmnaf/f mice. (H) Body weight of male LmnaG609G/G609G, LmnaG609G/+, Lmnaf/f;TC, and Lmnaf/f mice. *P < 0.05, Lmnaf/f;TC versus Lmnaf/f mice; ***P < 0.001, LmnaG609G/G609G versus Lmnaf/f mice. All data represent means SEM. P values were calculated by Students t test, except that statistical comparison of the survival data was performed by log-rank test. Photo credits: Weifeng Qin, Medical Research Center, Shenzhen University (A and B); Shimin Sun, School of Life Sciences, Shandong University of Technology; Medical Research Center, Shenzhen University (C).

Loss of Sirt7, an NAD+ (nicotinamide adenine dinucleotide)dependent deacylase, causes heart dysfunction with systemic inflammation and accelerates aging (22, 23). We noticed defective neovascularization in Sirt7 knockout mice (Fig. 5A). Knockdown of Sirt7 up-regulated the levels of interleukin-1 (IL-1) and IL6 in HUVECs, as determined by Western blotting and real-time PCR (Fig. 5, B and C). Significantly, the protein level of Sirt7 was reduced almost 50% in Lmnaf/f;TC MLECs (Fig. 5D). By contrast, the levels of Sirt6 and Sirt1 were hardly decreased in Lmnaf/f;TC MLECs. Furthermore, co-immunoprecipitation revealed that Lamin A interacted with Sirt7, which was significantly enhanced in the case of progerin (Fig. 5E). FLAG-SIRT7 was polyubiquitinated, which was enhanced in the presence of progerin compared with Lamin A (Fig. 5F). Ectopic expression of progerin in human embryonic kidney (HEK) 293 accelerated SIRT7 protein degradation, which was inhibited by MG132 (a proteasome inhibitor) (Fig. 5G). These data suggest that accumulation of progerin destabilizes Sirt7 by proteasomal pathway in progeria cells.

(A) Quantification of blood flow recovery following hindlimb ischemia in Sirt7/ and Sirt7+/+ mice. (B) Left: Representative immunoblots showing indicated protein levels in HUVECs treated with si-SIRT7 or scramble (Scram). Right: Quantification of relative protein levels. *P < 0.05 and **P < 0.01, small interfering RNA (siRNA) versus Scram. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. (C) Real-time PCR analysis of the indicated gene expression in HUVECs treated with si-SIRT7 or Scram. *P < 0.05, siRNA versus Scram. (D) Left: Representative immunoblots showing indicated sirtuin protein levels in FACS-sorted MLECs. Right: Quantification of relative protein levels. *P < 0.05. Note that down-regulated Sirt7 but rather up-regulated Sirt6 and hardly changed SIRT1 in Lmnaf/f;TC MLECs. (E) Left: Co-immunoprecipitation (IP) experiments showing hemagglutinin (HA)SIRT7 in antiFLAGLamin A and antiFLAG-progerin immunoprecipitates. Right: Quantification of relative protein levels. *P < 0.05. (F) Left: Representative immunoblots showing polyubiquitinated SIRT7, which was up-regulated in the presence of progerin but rather down-regulated in the presence of Lamin A. Right: Quantification of relative protein levels. *P < 0.05. (G) Representative immunoblots showing SIRT7 protein levels in the presence of Lamin A or progerin in HEK293 cells treated with cycloheximide (CHX) and/or MG132 (M). Quantification of relative SIRT7 protein levels was shown. *P < 0.05, progerin versus Lamin A. All data represent means SEM. P values were calculated by Students t test. Photo credit: Xiaolong Tang, Medical Research Center, Shenzhen University (B, D, E, F, and G).

We reasoned that Sirt7 might underlie the VE dysfunction in progeria mice. To test this hypothesis, we first examined whether ectopic Sirt7 could rescue the exacerbated inflammatory response in HUVECs. As shown, overexpression of SIRT7 significantly down-regulated the expression of multiple inflammatory genes such as IL1 (Fig. 6A). To test the in vivo function of Sirt7 in defective neovascularization, we generated a recombinant AAV serotype 1 (rAAV1) cassette with Sirt7 gene expression driven by a synthetic ICAM2 promoter (IS7O), which ensures VE-specific expression (24, 25). As shown, on-site injection of IS7O at a dose of 1.25 1010 viral genome-containing particles (vg)/50 l significantly improved blood vessel formation in Lmnaf/f;TC mice (Fig. 6B). The ectopic expression of Sirt7 and the increase in CD31-labeled ECs were evidenced by fluorescence confocal microscopy in ECs of regenerated blood vessels (Fig. 6, C and D).

(A) Real-time PCR analysis of genes that are aberrantly up-regulated in progerin-overexpressing HUVECs upon overexpression of SIRT7. *P < 0.05, **P < 0.01, and ***P < 0.001. (B) Neovascularization assay in Lmnaf/f;TC mice with hindlimb ischemia, treated with or without IS7O particles. **P < 0.01. (C) Immunofluorescence microscopy analysis of FLAG-SIRT7 and CD31 expression in gastrocnemius muscle 14 days after femoral artery ligation. Scale bar, 25 m. (D) Percent CD31+ ECs in Lmnaf/f;TC mice treated with or without IS7O particles. ***P < 0.001. (E) Representative immunofluorescence images of the liver, aorta, and muscle of Lmnaf/f;TC mice after IS7O therapy, showing CD31+ ECs with FLAG-SIRT7 expression. Scale bar, 50 m. (F) Representative immunoblots showing expression of FLAG-SIRT7 in aorta and WBMCs. Note that FLAG-SIRT7 was merely detected in WBMCs. (G) Life span of IS7O-treated and untreated Lmnaf/f;TC and LmnaG609G/+ mice. (H) Body weight of IS7O-treated and untreated Lmnaf/f;TC and Lmnaf/f mice. All data represent means SEM. P values were calculated by Students t test, except that the statistical comparison of survival data was performed by log-rank test. Photo credits: Shimin Sun, School of Life Sciences, Shandong University of Technology; Medical Research Center, Shenzhen University (C and E); Xiaolong Tang, Medical Research Center, Shenzhen University (F).

We next asked whether IS7O could ameliorate premature aging and extend life span. To this end, the IS7O particles were injected via tail vein from 21 weeks of age, when progeria mice start to die. The injection was repeated every other week at a concentration of 5 1010 vg/200 l per mouse. While all untreated mice died before 34 weeks of age, most IS7O-treated mice were still alive at the age of 44 weeks, when they were euthanized for histological analysis. The ectopic expression of FLAG-SIRT7 was observed in the ECs of liver, muscle, and aorta, but not in whole bone marrow cells (WBMCs), determined by fluorescence microscopy and/or Western blotting (Fig. 6, E and F). The median life span was extended by 76%from 25 to >44 weeks (Fig. 6G). The age-related body weight loss was slightly rescued upon IS7O therapy in Lmnaf/f;TC mice (Fig. 6H). These data suggest that progerin-caused VE dysfunction and systemic aging are partially, if not entirely, attributable to Sirt7 decline.

Mounting evidence supports the idea that endothelial dysfunction is a conspicuous marker for vascular aging and CVDs (2628). However, the fundamental question whether VE dysfunction causally triggers systemic aging remains. The heterogeneity of vascular cells and their close communication with the bloodstream render it difficult to understand the primary function of the VE. The murine LmnaG609G mutation, equivalent to the LMNAG608G found in humans with HGPS, causes premature aging phenotypes in various tissues and organs, thus providing an ideal model for studying aging mechanisms at both tissue and organismal levels. Data from the LmnaG609G model suggest that SMCs are the primary cause of vascular diseases, such as atherosclerosis (10, 11). A recent study showed that specific expression of LmnaG609G in SMCs causes atherosclerosis and shortens life span in atherosclerosis-prone Apoe/ mice (12). We used Tie2-Cre line to generate the VE-specific LmnaG609G mouse model. Lmnaf/f;TC mice exhibited vascular dysfunction, accelerated aging, and a shortened life span to a similar extent to the whole-body LmnaG609G model. Tie2 expression was reported not only in ECs but also in hematopoietic lineages (29). Our single-cell transcriptomic data identified Tie2 transcripts mainly in MLECs instead of B-, T-, or M-like cells. When a synthetic ICAM2 promoter was used to drive ectopic expression of FLAG-SIRT7 in the rescue experiments, ectopic FLAG-SIRT7 was successfully detected in ECs of the aorta, muscle, and liver but hardly detected in WBMCs. Therefore, Tie2-driven progerin expression combined with synthetic ICAM2-drivern SIRT7 rescue largely ensures the EC-specific contribution in systemic aging. Of note, although the number and function of hematopoietic stem cells decline in another progeria model, Zmpste24/ mice (30), little effect was observed when healthy hematopoietic progenitor cells were transplanted to Zmpste24/ mice in the context of systemic aging. Recently, Hamczyk et al. (12) found that knocking in the LmnaG609G allele in macrophages mediated by LysM-Cre merely affects aging and life span. Therefore, our data strongly suggest that, as the largest secretory organ (3), VE is pivotal in regulating systemic aging and longevity. In support of our findings, Foisner et al. (31) reported that VE-cadherin promoter-driven expression of progerin in a transgenic line causes cardiovascular abnormalities and shortens life span.

One limitation in the understanding of mechanisms of VE dysfunction is the vascular cell heterogeneity and the lack of appropriate in vitro system for ECs. Here, we took advantage of single-cell RNA sequencing technique to analyze the transcriptomes of MLECs. Unexpectedly, although >95% purity was achieved by FACS, MLECs isolated by CD31 immunofluorescence labeling turned out to be a mixture of cells, including ECs and T-, B-, and M-like cells. Although enriched by FACS, these non-ECs expressed low level of CD31 mRNA, raising the possibility that cell surface proteins such as CD31 T-, B-, and M-like cells might be obtained from neighbor ECs via intercellular protein transfer (32). Nevertheless, these findings suggest that one cannot just purify CD31+ cells and pool them together for mechanistic study, because one might arrive at a misleading conclusion. We compared the expression of genes that are associated with atherosclerosis, arthritis, heart failure, osteoporosis, or amyotrophy (the Online Mendelian Inheritance in Man; https://omim.org) between progeroid and control in all four clusters. An obvious alteration of these genes/pathways was observed mainly in ECs and M-like cells (fig. S2). At the current stage, it is hard to separate cell-autonomous and paracrine effects among different cell populations. In the future, it would be worthwhile to do an analysis in Lmnaf/f;TC MLECs. The data will be useful to study the paracrine effect of ECs on other cell populations.

Since the identification of the causal link between LMNA G608G mutation and HGPS, numerous efforts have been put on the development of treatment for HGPS. Farnesyltransferase inhibitors (33), resveratrol, and N-acetyl cysteine (30) treatment alleviate premature aging features and extend life span in progeria murine models. Rapamycin (34) and metformin (35) incubation rescue senescence in HGPS cells. On the basis of these notions, patients with HGPS taking a farnesyltransferase inhibitor, lonafarnib, in a clinical trial showed significant improvement of health status, reduction of mortality rate, and a potential extension of life span (about 1 to 2 years) (36). Taking advantage of gene therapy and the dispensable role of Lamin A, morpholino oligos (9), and CRISPR-Cas9 designs (37, 38), which prevent Lamin A/progerin generation, can alleviate aging features and extend life span from 25 to 40% in progeria mice. However, considering the indispensable function of Lamin A in humans, these genome-modifying strategies need further experimentation before potential clinical application. Here, applying a different strategy, we showed that rAAV1-SIRT7 (IS7O), targeting dysfunctional VE, largely ameliorates progeroid features and almost doubles the median life span (from 25 to >44 weeks). To our best knowledge, this is the most marked rescue of progeria in a mouse model via gene therapy. Given that SIRT7 elicits deacylase activity to modulate cellular functions (22, 23), it is worthwhile to identify small molecules that specifically target SIRT7 activity for therapeutics in the future. Resveratrol is a potential activator of SIRT1, as well as SIRT7 (39), and has protective effects on vascular function and blood pressure (40). Further depicting the relationship of SIRT7 and resveratrol in the regulation of vascular function would help in seeking leading compounds of SIRT7 specific activators.

Collectively, we reveal VE dysfunction as a primary trigger of systemic aging and as a risk factor for age-related diseases such as atherosclerosis, heart failure, and osteoporosis. Drugs and molecules that target VE might serve as good candidates in the treatment of age-related diseases other than CVDs. The findings in SIRT7-based gene therapy implicate great clinical potentials for progeria as well as in antiaging applications.

Lmnaf/+ allele (LmnaG609G mutation flanked by two loxP sites) was generated by Cyagen Biosciences Inc., China. Briefly, the 5 and 3 homology arms were amplified from bacterial artificial chromosome clones RP23-21K15 and RP23-174J9, respectively. The G609G (GGC to GGT) mutation was introduced into exon 11 in the 3 homology arm. C57BL/6 embryonic stem cells were used for gene targeting. To obtain ubiquitous expression of progerin (LmnaG609G/G609G), Lmnaf/f mice were bred with E2A-Cre mice. To obtain VE-specific expression of progerin, Lmnaf/f mice were bred with Tie2-cre mice. Mice were housed and handled in accordance with protocols approved by the Committee on the Use of Live Animals in Teaching and Research of Shenzhen University, China.

Four-month-old male mice were anesthetized with 4% chloral hydrate (0.20 ml/20 g) by intraperitoneal injection. Hindlimb ischemia was performed by unilateral femoral artery ligation and excision, as previously described (41). In brief, the neurovascular pedicle was visualized under a light microscope following a 1-cm incision in the skin of the left hindlimb. Ligations were made in the left femoral artery proximal to the superficial epigastric artery branch and anterior to the saphenous artery. Then, the femoral artery and the attached branches between ligations were excised. The skin was closed using a 4-0 suture line, and erythromycin ointment was applied to prevent wound infection after surgery. Recovery of the blood flow was evaluated before and after surgery using a dynamic microcirculation imaging system (Teksqray, Shenzhen, China). Relative blood flow recovery is expressed as the ischemia-to-nonischemia ratio. At least three mice were included in each experimental group.

HEK293 cells and HUVECs were purchased from the American Type Culture Collection. HEK293 cells were cultured in Gibco Dulbeccos modified Eagles medium (Life Technologies, USA) supplemented with 10% fetal bovine serum at 37C, 5% CO2. HUVECs were cultured in Gibco M199 (Life Technologies, USA) supplemented with 15% fetal bovine serum, EC growth supplement (50 g/ml), and heparin (100 g/ml) at 37C, 5% CO2. All cell lines used were authenticated by short tandem repeat profile analysis and were mycoplasma free.

Total RNA was extracted from cells or mouse tissues using TRIzol reagent RNAiso Plus (Takara, Japan) and transcribed into complementary DNA (cDNA) using 5 PrimeScript RT Master Mix (Takara, Japan), following the manufacturers instructions. The mRNA levels were determined by quantitative PCR with SYBR Premix Ex Taq II (Takara, Japan) detected on a CFX Connect Real-Time PCR Detection System (Bio-Rad). All primer sequences are listed in table S1.

For protein extraction, cells were suspended in SDS lysis buffer and boiled. Then, the lysate was centrifuged at 12,000g for 2 min, and the supernatant was collected. For Western blotting, protein samples were separated on SDS-polyacrylamide gels, transferred to polyvinylidene difluoride membranes (Millipore, USA), blocked with 5% nonfat milk, and incubated with the relevant antibodies. Images were acquired on a Bio-Rad system. All antibodies are listed in table S2.

Frozen sections of aorta, skeletal muscle, and liver tissues were fixed in 4% paraformaldehyde (PFA), permeabilized with 0.3% Triton X-100, blocked with 5% bovine serum albumin and 1% goat serum, and then incubated with primary antibodies at room temperature for 2 hours or at 4C overnight. After three washes with phosphate-buffered saline with Tween 20, the sections were incubated with secondary antibodies for 1 hour at room temperature and then stained with 4,6-diamidino-2-phenylindole antifade mounting medium. Images were captured under a Zeiss LSM 880 confocal microscope. All antibodies are listed in table S2.

Paraffin-embedded sections of PFA-fixed tissues were dewaxed and hydrated. Staining was then performed using a Masson trichrome staining kit (Beyotime, China). In brief, the sections were dipped in Bouin buffer for 2 hours at 37C and then successively stained with Celestine blue staining solution, hematoxylin staining solution, Ponceau S staining solution, and aniline blue solution for 3 min. After dehydrating with ethyl alcohol three times, the sections were mounted with Neutral Balsam Mounting Medium (BBI Life Science, China). Images were captured under a Zeiss LSM 880 confocal microscope.

Mice were euthanized by decapitation. The lungs were then collected, cut into small pieces, and then digested with collagenase I (200 U/ml) and neutral protease (0.565 mg/ml) for 1 hour at 37C. The isolated cells were incubated with phycoerythrin-conjugated anti-CD31 antibody for 1 hour at 4C and then 7-aminoactinomycin D (7-AAD) (1:100) for 5 min. CD31-positive and 7-AADnegative cells were sorted on a flow cytometer (BD Biosciences, USA).

Four-month-old male mice were anesthetized with 4% chloral hydrate by intraperitoneal injection. Thoracic aortas were collected, rinsed in ice-cold Krebs solution, and cut into 2-mm-length rings. Each aorta ring was bathed in 5-ml oxygenated (95% O2 and 5% CO2) Krebs solution at 37C for 30 min in a myograph chamber (620M, Danish Myo Technology). Each ring was stretched in a stepwise fashion to the optimal resting tension (thoracic aortas to ~9 mN) and equilibrated for 30 min. Then, 100 mM K+ Krebs solution was added to the chambers to elicit a reference contraction and then washed out with Krebs solution at 37C until a baseline was achieved. Vasodilation induced by Ach or SNP (1 nM to 100 M) was recorded in 5-hydroxytryptamine (2 M) contracted rings. Data are represented as a percentage of force reduction and the peak of K+-induced contraction. At least three mice were included in each experimental group.

Seven- to 8-month-old male mice were anesthetized by isoflurane gas inhalation and then subjected to transthoracic echocardiography (iU22, Philips). Parameters, including heart rate, cardiac output, left ventricular posterior wall dimension, left ventricular end-diastolic dimension, left ventricular end-systolic diameter, LV ejection fraction, and LV fractional shortening, were acquired. At least three mice were included in each experimental group.

Seven- to 8-month-old male mice were euthanized by decapitation. The thigh bone was fixed in 4% PFA at 4C overnight. The relevant data were collected by micro-CT (Scanco Medical, CT100). At least three mice were included in each experimental group.

A Rota-Rod Treadmill (YLS-4C, Jinan Yiyan Scientific Research Company, China) was used to monitor fatigue resistance. Briefly, mice were placed on the rotating lane, and the speed of the rotations gradually increased to 40 rpm. When the mice were exhausted, they were safely dropped from the rotating lane, and the latency to fall was recorded. At least three mice were included in each experimental group.

CD31+ cells isolated from murine lung by FACS (>90% viability) were used for single-cell RNA sequencing. A sequence library was built according to the Chromium Single-Cell Instrument library protocol (42). Briefly, single-cell RNAs were barcoded and reverse-transcribed using the Chromium Single-Cell 3 Reagent Kits v2 (10 Genomics) and then fragmented and amplified to generate cDNAs. The cDNAs were quantified using an Agilent Bioanalyzer 2100 DNA Chip, and the library was sequenced using an Illumina Hiseq PE150 with ~10 to 30M raw data assigned for each cell. The reads were mapped to the mouse mm9 genome and analyzed using STAR: >90% reads mapped confidently to genomic regions and >50% mapped to exonic regions. Cell Ranger 2.1.0 was used to align reads, generate feature-barcode matrices, and perform clustering and gene expression analysis. Mean reads (>80,000) and 900 median genes per cell were obtained. The unique molecular identifier counts were used to quantify the gene expression levels, and the t-distributed stochastic neighbor embedding (t-SNE) algorithm was used for dimensionality reduction. The cell population was then clustered by k-means clustering (k = 4). The Log2FoldChange was the ratio of gene expression of one cluster to that of all other cells. The P value was calculated using the negative binomial test, and the false discovery rate was determined by the Benjamini-Hochberg procedure. GO and KEGG enrichment analyses were performed in DAVID version 6.8 (43).

A two-tailed Students t test was used to determine statistical significance, except that the statistical comparison of survival data was performed by log-rank test. All data are presented as the means SD or means SEM, as indicated, and a P value <0.05 was considered statistically significant.

Supplementary material for this article is available at http://advances.sciencemag.org/cgi/content/full/6/8/eaay5556/DC1

Fig. S1. Generation of Lmnaf/f mice and phenotypic analysis of LmnaG609G/G609G mice.

Fig. S2. Single-cell transcriptomic analysis of CD31+ MLECs.

Fig. S3. VE-specific progerin expression.

Fig. S4. Vasodilation analysis of LmnaG609G/+ mice.

Fig. S5. Expression of atherosclerosis- and osteoporosis-associated genes in MLEC transcriptomes.

Table S1. List of primer sequences.

Table S2. List of antibodies.

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial license, which permits use, distribution, and reproduction in any medium, so long as the resultant use is not for commercial advantage and provided the original work is properly cited.

Acknowledgments: We thank J. Tamanini (Shenzhen University and ETediting) for editing the manuscript before submission. Funding: This study was supported by grants from the National Natural Science Foundation of China (91849208, 81571374, 91439133, 81871114, 81601215, 81972602, and 81702909), the National Key R&D Program of China (2017YFA0503900), the Science and Technology Program of Guangdong Province (2014A030308011, 2017B030301016, and 2019B030301009), and the Shenzhen Municipal Commission of Science and Technology Innovation (JCYJ20160226191451487, KQJSCX20180328093403969, JCYJ20180507182044945, ZDSYS20190902093401689, and Discipline Construction Funding of Shenzhen 2016-1452). Author contributions: B.L. designed and supervised the project. S.S., W.Q., and X.T. conducted experiments with help from W.H., S.Z., M.Q., Z.L., X.C., Q.P., and B.Z. Y.M. performed bioinformatic analysis. Z.W. and Z.Z. provided resources. S.S., X.T., and B.L. wrote the manuscript. All authors discussed the experimental results and reviewed 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. The data of single-cell transcriptomics are available in the GEO database (GSE138975). Additional data related to this paper may be requested from the authors.

See the article here:
Vascular endotheliumtargeted Sirt7 gene therapy rejuvenates blood vessels and extends life span in a Hutchinson-Gilford progeria model - Science...

The fight against blood cancer continues – Sherbrooke Record

Calling all hockey fans. The Hockey Fights Cancer fundraiser for Maisonneuve Rosemont Hospital will take place this Sunday, Feb. 23 at 3:30 p.m. in the Bishops arena. Four teams of friends, firefighters and plain old hockey fanatics will take the ice to help fund cutting edge research in stem cell therapy to treat blood cancer. The tournament is free and open to the public, and anyone interested in supporting the cause or just watching the games is welcome to attend. The tournament started last year after local firefighter Eric Mackeage, diagnosed with a rare form of blood cancer, received treatment at the Maisonneuve Rosemont Hospital. When friends of Mackeages stepped up wanting to help, it was decided the best way would be to raise money to support the research that Mackeage believes saved his life. Im an experiment, Mackeage said. Firefighters are at an increased risk of developing the type of cancer Mackeage was diagnosed with. See full story in the Thursday, Feb. 20 edition of The Record.

Read more:
The fight against blood cancer continues - Sherbrooke Record

Survivor Legend Ethan Zohn Discusses His Journey Through Cancer and Back to the Island – Parade

This past Wednesday, millions of Survivor fans got to see 20 previous winners from the reality shows storied past return for another shot at victory and a $2 million prize. And while each of them had a journey to getting back to the island, season 3 winner Ethan Zohn may have the most inspirational story of them all. Because up until seven years ago, not only was it up in the air whether Ethan would be playing Survivor again in 2020, it was unlikely he would even be alive to see season 40.

In 2009, five years after his last Survivor appearance, Ethan was diagnosed with a rare type of cancer called CD20-positive Hodgkins lymphoma. He underwent several treatments since his diagnosis and was declared cancer-free in late April 2010. Then, after nearly 20 months of remission, cancer returned in his chest. Not to be deterred, Ethan continued to undergo stem-cell transplants, all the while doing charity work to promote his cause, as well as his charity Grassroot Soccer, which uses soccer to raise money and awareness to fight HIV/AIDS. In March 2013, Ethan officially declared he was cancer-free once more and has been in remission ever since. With a clean bill of health, hes now ready to return to the game that changed him forever, with a new outlook on life.

Ethan talks with Parade.com about the experience of engaging with both Survivor and the fandom after more than a decade away, the struggles he underwent after becoming cancer-free, and his decision to use his time in the spotlight to give back.

Related: Survivor Season 40: Everything We Know So Far (Including Who Was Voted Out)!

This time last week, you got to watch your return with a special screening of the premiere on the big screen in LA What was that experience like, the culmination of everything you went through to this point?Watching myself, I cant believe they make us run around in our underwear! Ill just put that out there. Im like, Jeff, they know were not shipwrecked. We get it. Give us some bathing suits, buddy! Nobody wants to watch anyones junk flop around. (Laughs.)

But watching myself on TV was a rush. Its a snapshot of my life. Its my highlight reel, and this is my song. Being here is just another chapter in this incredibly blessed life that Survivor has given me. Ive never watched the show with such a live audience, with fans cheering for different people and moves. It heightens the excitement for me. I realize how lucky I am and the moment we have right here. Im just trying to drink it all in and have so much fun.

Whats it been like to engage with the fan community, considering how different it looks between now and when you first played?When we played the game, it was like we were playing underwater compared to now. We had no idols, no clues, no ways to get back into the game, nothing. To take that one step further, we had no social media. We had printed media, radio, maybe some television spots. The way in which information is passed around is like light speed. That is indicative of the way the world works and how the games played. We delved deep into personal relationships, honor, integrity, trust, teamwork, and survival. We depended on each other to survive daily out there in Africa. Now its like boom, boom, blindsides, backstabs, idol clues. Its fast-paced, rocket fuel injected into the game of Survivor both inside and outside the show.

For me, just trying to keep up with social media is a thing in and of itself! (Laughs.) Im trying to figure out whats going on right now. But its been fun to interact with it because, surprisingly, a lot of fans Ive been connecting with are young. They were either extremely young or not alive when I was on the show. So to see a new generation of fans getting excited about the greatest show on earth is pretty exciting.

Youve been considered one of the most well-liked and revered players in Survivor history. What has been your reaction to that label?It feels wonderful, to be honest. (Laughs.) It lets me reminisce about my past in a way thats really fun and exciting for my family and me. I definitely didnt drop off the face of the earth, but Ive been in the public eye recently more as a cancer survivor than a Survivor player. So its really fun and exciting. Im very appreciative of what this show has enabled me to do in my life. So coming back on was wonderful. Its also paying tribute to the life the show has given me, to everyone who has come before and after me who made this show what it is today.

I specifically remember the first second of the game, when we lined up on the beach. I looked down and saw $21 millionbecause Sandras won twiceas well as a history lesson. Youre looking at each person and seeing 20 years of my life, of Jeffs life, of everyones lives. Its rare that any TV show has the shelf life Survivor has. I remember in season 3, people say, Oh, this is the last season. Theres no way this is going to go another season. (Laughs.) September 11th happened, and nobody wanted to see a show called Survivor and see people suffer. Now look at us, 20 years later!

Though youve been away from Survivor for a while, you certainly havent strayed away from reality television at large. What drove you to want to appear on other shows, even if they werent on an island?Right place, right time. It was the start of reality television as a genre. All these new shows were coming out, and having been on Survivor, most of the time, they were celebrity reality shows. I just said yes to everything. I never thought it would last that long. So I thought, Im going to get in and have as much fun as I possibly can. Because I never know when this is going to end. So I said yes to every single opportunity possible. Why not?! (Laughs.) Plus, its always a good opportunity for me. I could wear a Grassroot Soocer t-shirt, or talk about my experience with cancer.

Its good to have something valuable in your life that isnt reality TV. I went on reality TV because it was a crazy adventure. I wanted to compete at the highest level in front of millions of people. Youre never going to get that anywhere. I didnt grow up watching Survivor dreaming of being on, like an Adam Klein. People like him, Michele, Wendell, and Ben grew up on when I was on the show, and here they are coming out to play again because its been their lifelong dream to be on Surivor. My lifelong dream was to be a professional soccer player. Once I accomplished that, this was the icing on the cake.

Its interesting you talk about never knowing when your reality TV career would end, given the way your life went. I cant imagine what your first thought was when you received your initial cancer diagnosisGetting diagnosed as a young adult at 35 years with a rare form of blood cancer was completely shocking to me. My only connection to cancer was through my father, who passed away when I was 14. So I only thought, Cancer = death. I was completely frightened and confused, as you can imagine. At that point, all my other friends were just starting their lives. They were beginning jobs and families, moving to new homes and towns. I was forced to press pause on my life, while everyone else was starting theirs. That was difficult for me. But coming out of the cancer diagnosis was more difficult for me personally.

Related: Survivor Winners at War: Lex van den Berghe on Amber Mariano and Old School Tactics

Can you elaborate on that?When you get diagnosed, and the doctor tells you to do something or youll die, you do it. Theres no choice; its not that hard. But then, when youre deemed healthy and in remission, all the doctors and nurses go away. And your friends start to pull away. Thats when it really got hard. Dealing with the anxiety and the fear of relapse, the dump trucks full of uncertainty, the scars that need healing. Now Im a young adult with no job. Im infertile; whos going to marry me? Is it going to come back? Theres all this stuff in your mind as a young man that an older adult doesnt face. I had to get to a point in my life where I was mentally clear, physically strong enough, and spiritually open to having an experience like Survivor. Thats the win for me. Just getting to the starting line is probably harder than anything Ill have to do for the rest of my life.

Obviously Survivor and fighting cancer are like apples and orange. But is there anything you pulled from that experience mentally or emotionally that helped you during your treatments?I do think my time on Survivor enabled me to learn how far I can push my body. Mentally, physically, spiritually, environmentally, socially. On Survivor, you push yourself to the max, and then you have to go even further. Coming into cancer, Im like, Okay, Ive suffered before. I know this is going to suck, but Ive been here before. Youve just got to take it day by day and minute by minute. You never know whats around the corner, whether it be an Immunity Challenge or a clinical trial.

Survivor is a game of relationships. Youve got to make friends with these people, but friendship is based on trust, and you cant really trust anyone. With cancer, you are surrounded by so many people who love you. But it is a lonely feeling. No one knows what its like to have cancer unless youve been through it yourself. Even then, everyones experience is different based on who you are. There is loneliness out there on Survivor. Even though you have a tribe and alliances, the result is having fewer and fewer people around you. The concept of community exists at a level. But in the back of everyones minds, they want you dead. (Laughs.) So does cancer. There are surprisingly some parallels.

When you get the call to go out for season 40, what went through your head, considering how much your life has changed from the last time you played?The game has changed, and so have I. (Laughs.) Its not better or worse now; its just different. Any good player in any sport has to adapt and be open to playing with the rules that exist for that specific game at that moment. I had gotten to a point where I was comfortable in life, with my wife and my two cats in New Hampshire. Im not the 27-year-old who doesnt care what happens to him anymore. Im now playing with the fact that Ive had to go through cancer and stem cell treatment twice. No one else has brought that kind of baggage into the game. Yes, everyone out there has a story and challenges weve overcome to get to where we are. I dont think anyone has had the health challenges that Ive had going out there. I was a little nervous about leaving the world I lived in to play that game, to be honest.

Once you made that decision, how did you make sure you were ready to go back into the game?I knew I had to do some catching up! I knew I had to get off the couch and stop smoking weed; cannabis and CBD have been huge in helping my post-cancer anxiety medically speaking. I left New Hampshire, because they called me in January and I knew I couldnt swim in a frozen lake. My wife and I moved down to Atlanta and I started training outside. I was swimming, doing puzzles, reading body language and lip reading books. I met with my shrink; my wife hid idols in the forest for me to find every day. I thought, If Im going to do this, to ease my fears that Im an old-schooler who has no place in the modern game, I want to get ready.

Related: Survivor Winners at War: Reed Kelly Talks Natalie Anderson and the Power of Partnerships

What role has the show served in your life, especially as the years passed between appearances? I know youve told the story about receiving a stem cell treatment while you were watching the Heroes vs. Villains premiere.Survivor is a wonderful word to describe me for a lot of different reasons. Its been a constant in my life. Im still known as the Survivor guy. I do a lot of speaking engagements, and my notoriety comes from the fact that I was on Survivor. Im not trying to hide that fact. Survivor changed my life. It sent me on a trajectory and enabled me to do so many things I would have dreamed of doing. I am incredibly grateful and appreciative of what this show has offered me.

Not only is the show creating fans around the world, but they are also saving lives. This show is completely responsible for the fact that I was able to co-found Grassroot Soccer. I played professional soccer in Zimbabwe leading up to my time on Survivor: Africa. During the show, I played hackeysack with the children in a local village. It was a real-life moment in the middle of this cutthroat game. When I won the money, I wanted to do something great with that. I met up with some buddies of mine, and we co-founded Grassroot Soccer. Were an adolescent health organization that has programs running in 63 countries and graduated 2.3 million kids, all because of Survivor.

Let me take that one step further for you. When I got cancer, we had just lost [Palau contestant] Jennifer Lyon. We were battling at the same time, and she didnt make it, unfortunately. In response to that, I created Survivor Stand Up to Cancer, which was a partnership between the show and the cancer research organization. Every year, props are auctioned off, and the proceeds go to cancer research. Some of the money raised from 2009 to 2012 was used to fund an experimental new drug that saved my life. Talk about full circle! This show is so much bigger than people imagine, especially in my life.

You get the chance to come back to the show that has meant so much to you. Your feet hit the sands of Fiji, and you realize youre playing Survivor again. Describe that feeling for me.Just thinking about it gives me chills. Theres so much emotion. My heart was pumping; my brain was going crazy. Theres a lot of pressure that Im putting on myself, the pressure to perform well, make alliances, not look stupid, and win. All this stuff is going through your head at that exact moment. The games about to start; Jeff Probsts waiting for you. Youre just walking into one of the most uncomfortable comfortable situations. Ive dreamed of this moment, hoping it unfolds positively. It was emotional. Its a perfect little bow on this amazing life that Survivor has given me.

Celebrity interviews, recipes and health tips delivered to yourinbox.

See the rest here:
Survivor Legend Ethan Zohn Discusses His Journey Through Cancer and Back to the Island - Parade

FDA Scoffs at Third Way Forward in Stem Cell Therapy – Pain News Network

Gimble and his co-authors recommend the FDA re-evaluate how it categorizes tissues as structural or cellular to recognize the different safety profiles of stem cells products. They also think the FDA should work with accreditation agencies like AABB and FACT to develop meaningful accreditation standards, along with a national registry for stem cell therapies.

This measured third way seeks to carve a compromise between the FDAs regulationist faction and wild west stem cell providers -- a new ideological center that synthesizes the aspirations of two opposing parties in an effort to achieve a compromise.

It must be stated that there are serious questions as to whether the authors proposed polarity is in fact an artifice created for the specific purpose of legitimizing their third way. Upon serious inspection, the authors stated dangers of stem cell clinics may actually be a disingenuous straw man created for their own business interests.

To promote their own agenda and to gain favor with the FDA, Gimble and his co-authors seem to have thrown stem cell clinicians like Dr. Mark Berman under the proverbial bus. Berman, who is a defendant in a FDA lawsuit over his use of autologous cells, recently won a victory in federal court. The judge found that the FDA may not have regulatory authority over Bermans procedures and that a trial needs to be held to resolve the issue.

Nevertheless, the impetus behind the authors recommendations is to move forward with bringing stem cells to patients faster and in a safer manner. Regrettably, the FDA does anything but take the authors seriously. In a lengthy response to the Gimble article, Dr. Peter Marks, Director of the FDAs Center for Biologics Evaluation and Research, merely reiterates the agencys firmly-established regulationist position.

After commending the authors for their desire to accelerate the scientific investigation and development of stem cell therapies, Marks demonstrates the FDAs backward-looking posture by stating the agencys regulation of stem cells is distinct from the practice of medicine and should be left alone.

This is an existing paradigm that has been in place for decades, Marks wrote. Autologous cellular therapies do hold tremendous promise, but they will only find their way into routine clinical practice to bring benefit to all patients if they are held to the same standards to demonstrate safety and efficacy as other unproven medical products.

Marks attempts to bolster his argument by citing patient safety, the dearth of research on adipose-derived stem cells and the unethical bad actor clinics that exploit desperate patients. However, the spirit of his position reveals a resistance to any sort of change whatsoever.

Marks and the FDA are living in the past. They consider your cells to be unproven medical products. Apparently, they have yet to realize that the stem cell poles have already shifted.

A. Rahman Ford, PhD, is a lawyer and research professional. He is a graduate of Rutgers University and the Howard University School of Law, where he served as Editor-in-Chief of the Howard Law Journal. Rahman lives with chronic inflammation in his digestive tract and is unable to eat solid food. He has received stem cell treatmentin China.

Visit link:
FDA Scoffs at Third Way Forward in Stem Cell Therapy - Pain News Network