Page 533«..1020..532533534535..540550..»

Stem Cell and Gene Therapy Biological Testing Market Future Developments, Business Insights, End Users, Application and Forecast to 2029 – Digital…

Posted: January 20, 2022 at 2:20 am

Stem Cell and Gene Therapy Biological Testing Market research report are based upon SWOT analysis on which businesses can rely confidently. With the precise and high-tech information about healthcare industry, businesses can know about the types of consumers, consumers demands and preferences, their perspectives about the product, their buying intentions, their response to particular product, and their varying tastes about the specific product already existing in the market through this report. This information and market insights covered in the superior Stem Cell and Gene Therapy Biological Testing market document assists with maximizing or minimizing the production of goods depending on the conditions of demand.

Strategically analyzed facts and figures of the market and keen business insights covered in the credible Stem Cell and Gene Therapy Biological Testing business document would be a major aspect in achieving enduring business growth. The report offers steadfast knowledge and information of revolutionizing market landscape, what already exists in the market, future trends or what the market expects, the competitive environment, and strategies to plan to outshine the competitors. This worldwide Stem Cell and Gene Therapy Biological Testing market research report finds out the general market conditions, trends, inclinations, key players, opportunities, geographical analysis and many other parameters that helps drive business into the right direction.

Download Sample of this Report to understand structure of the complete report (Including Full TOC, Table & Figures) @ https://www.databridgemarketresearch.com/request-a-sample/?dbmr=global-stem-cell-and-gene-therapy-biological-testing-market

Key Market Players mentioned in this report:MEDIPOSTSmith & NephewANTEROGEN.CO.,LTDPHARMICELL Co., LtdJCR Pharmaceuticals Co., LtdNuVasive, IncGilead Sciences, IncDendreon Pharmaceuticals LLCOrganogenesis IncOsirisSTEMCELL Technologies

Stem Cell and Gene Therapy Biological Testing Market Segmentation:-

By Types:Cell Therapy and Gene Therapy

By Application:Hospitals, Wound Care Centres, Cancer Care Centres, Ambulatory Surgical Centres and Others

Market Analysis and Insights: Global Stem Cell and Gene Therapy Biological Testing Market

Data Bridge Market Research analyses that the stem cell and gene therapy biological testing market will exhibit a CAGR of around 14.87% for the forecast period of 2022-2029. Rising approvals of GMP-certified facilities to manufacture stem cells, rising stem cell research activities and increasing public and private expenditure for the development of healthcare infrastructure especially in emerging economies are the major factors attributable to the growth of stem cell and gene therapy biological testing market. This signifies that the stem cell and gene therapy biological testing market value, which was USD 1,497.03 million in 2021, will rocket up to USD 4,538.22 million by the year 2029.

Browse Full Report Along With Facts and Figures @ https://www.databridgemarketresearch.com/reports/global-stem-cell-and-gene-therapy-biological-testing-market

Global Stem Cell and Gene Therapy Biological Testing Market Scope and Market Size

The stem cell and gene therapy biological testing market is segmented on the basis of product type and end users. The growth amongst these segments will help you analyse meagre growth segments in the industries, and provide the users with valuable market overview and market insights to help them in making strategic decisions for identification of core market applications.

By product type, the global stem cell and gene therapy biological testing market is segmented into cell therapy and gene therapy.

Stem Cell and Gene Therapy Biological Testing Market, By Region:

The stem cell and gene therapy biological testing market is analysed and market size insights and trends are provided by product type and end users as referenced above.

The countries covered in the stem cell and gene therapy biological testing market report are U.S., Canada and Mexico in North America, Germany, France, U.K., Netherlands, Switzerland, Belgium, Russia, Italy, Spain, Turkey, Rest of Europe in Europe, China, Japan, India, South Korea, Singapore, Malaysia, Australia, Thailand, Indonesia, Philippines, Rest of Asia-Pacific (APAC) in the Asia-Pacific (APAC), Saudi Arabia, U.A.E, South Africa, Egypt, Israel, Rest of Middle East and Africa (MEA) as a part of Middle East and Africa (MEA), Brazil, Argentina and Rest of South America as part of South America.

Table of Contents: Global Stem Cell and Gene Therapy Biological Testing Market

1 Introduction2 Market Segmentation3 Executive Summary4 Premium Insight5 Market Overview6 Covid-19 Impact on Stem Cell and Gene Therapy Biological Testing in Healthcare Industry7 Global Stem Cell and Gene Therapy Biological Testing Market, by Product Type8 Global Stem Cell and Gene Therapy Biological Testing Market, by Modality9 Global Stem Cell and Gene Therapy Biological Testing Market, by Type10 Global Stem Cell and Gene Therapy Biological Testing Market, by Mode11 Global Stem Cell and Gene Therapy Biological Testing Market, by End User12 Global Stem Cell and Gene Therapy Biological Testing Market, by Geography13 Global Stem Cell and Gene Therapy Biological Testing Market, Company Landscape14 Swot Analysis15 Company Profiles16 Questionnaire17 Related Reports

Get Full Table of Contents with Charts, Figures & Tables @ https://www.databridgemarketresearch.com/toc/?dbmr=global-stem-cell-and-gene-therapy-biological-testing-market

The research provides answers to the following key questions:

What is the estimated growth rate of the market for the forecast period 20222029? What will be the market size during the estimated period? What are the key driving forces responsible for shaping the fate of the Energy Harvesting System market during the forecast period? Who are the major market vendors and what are the winning strategies that have helped them occupy a strong foothold in the Energy Harvesting System market? What are the prominent market trends influencing the development of the Energy Harvesting System market across different regions? What are the major threats and challenges likely to act as a barrier in the growth of the Energy Harvesting System market? What are the major opportunities the market leaders can rely on to gain success and profitability?

The key questions answered in Stem Cell and Gene Therapy Biological Testing Market report are:

What are the market opportunities, market risks, and market overviews of the Stem Cell and Gene Therapy Biological Testing Market?

Inquire Before Buying This Research Report@ https://www.databridgemarketresearch.com/toc/?dbmr=global-stem-cell-and-gene-therapy-biological-testing-market

About Us:

Data Bridge Market Researchset forth itself as an unconventional and neoteric Market research and consulting firm with unparalleled level of resilience and integrated approaches. We are determined to unearth the best market opportunities and foster efficient information for your business to thrive in the market. Data Bridge Market Research provides appropriate solutions to the complex business challenges and initiates an effortless decision-making process.

Data Bridge adepts in creating satisfied clients who reckon upon our services and rely on our hard work with certitude. Get Customization and Discount on Report by emailing sopan.gedam@databridgemarketresearch.com . We are content with our glorious 99.9 % client satisfying rate.

Contact Us:

US: +1 888 387 2818

UK: +44 208 089 1725

Hong Kong: +852 8192 7475

Email corporatesales@databridgemarketresearch.com

Read more:
Stem Cell and Gene Therapy Biological Testing Market Future Developments, Business Insights, End Users, Application and Forecast to 2029 - Digital...

Posted in Stem Cell Therapy | Comments Off on Stem Cell and Gene Therapy Biological Testing Market Future Developments, Business Insights, End Users, Application and Forecast to 2029 – Digital…

Can Multiple Myeloma Patients Achieve a Durable Remission After Induction Therapy & Skip or Delay a Stem Cell Transplant? – SurvivorNet

Posted: January 20, 2022 at 2:20 am

To Skip or Delay a Stem Cell Transplant

A stem cell transplant may be the best treatment option for many patients with multiple myeloma. But is it always the answer? And can you put off the procedure?

First off, its important to note that not all multiple myeloma patients are eligible for a stem cell transplant. Factors that can impact a persons eligibility include age, fitness and co-morbidities (other current medical problems) such as heart, lung, kidney or liver problems. But even if youre unable to have a stem cell transplant, that doesnt mean your treatment wont be as effective as others who are eligible.

In a conversation with SurvivorNet, Dr. Jing Ye, a hematologist and oncologist at the University of Michigan Medicine, discusses the possibility of patients achieving lasting remission without undergoing a stem cell transplant. Multiple myeloma patients who are not eligible for a stem cell transplant typically have a prolonged period of induction treatment followed by maintenance therapy.

According to Dr. Ye, clinical trial data has shown that these patients can enjoy the same progression-free survival.

This has also opened up the conversation surrounding timing of the stem cell transplant for those who are eligible. In fact, some myeloma specialists think that a stem cell transplant should be performed right after induction treatment, but others think its OK to collect the stem cells and save them for actual implementation later on when the disease has relapsed.

Its interesting that the transplant actually is a treatment option developed quite some time ago, several decades ago, Dr. Ye says. Nowadays we have more and more chemo-free types of multiple myeloma treatment available. So there is also a debate in our myeloma field among experts (about when to offer the stem cell transplant).

And many multiple myeloma patients actually do try to save the treatment for relapse, according to Dr. Ye.

Patients now have an option if they would like to consider transplant at a later stage of their disease, she says.

Like the other variable aspects of multiple myeloma treatment, these different approaches will suit different people with different goals and circumstances. All of these options should be weighed with the close guidance of a multiple myeloma specialist.

Learn more about SurvivorNet's rigorous medical review process.

Joe Kerwin is a writer and researcher at SurvivorNet, based in New York City. Read More

First off, its important to note that not all multiple myeloma patients are eligible for a stem cell transplant. Factors that can impact a persons eligibility include age, fitness and co-morbidities (other current medical problems) such as heart, lung, kidney or liver problems. But even if youre unable to have a stem cell transplant, that doesnt mean your treatment wont be as effective as others who are eligible.

According to Dr. Ye, clinical trial data has shown that these patients can enjoy the same progression-free survival.

This has also opened up the conversation surrounding timing of the stem cell transplant for those who are eligible. In fact, some myeloma specialists think that a stem cell transplant should be performed right after induction treatment, but others think its OK to collect the stem cells and save them for actual implementation later on when the disease has relapsed.

Its interesting that the transplant actually is a treatment option developed quite some time ago, several decades ago, Dr. Ye says. Nowadays we have more and more chemo-free types of multiple myeloma treatment available. So there is also a debate in our myeloma field among experts (about when to offer the stem cell transplant).

And many multiple myeloma patients actually do try to save the treatment for relapse, according to Dr. Ye.

Patients now have an option if they would like to consider transplant at a later stage of their disease, she says.

Like the other variable aspects of multiple myeloma treatment, these different approaches will suit different people with different goals and circumstances. All of these options should be weighed with the close guidance of a multiple myeloma specialist.

Learn more about SurvivorNet's rigorous medical review process.

Joe Kerwin is a writer and researcher at SurvivorNet, based in New York City. Read More

Original post:
Can Multiple Myeloma Patients Achieve a Durable Remission After Induction Therapy & Skip or Delay a Stem Cell Transplant? - SurvivorNet

Posted in Stem Cell Therapy | Comments Off on Can Multiple Myeloma Patients Achieve a Durable Remission After Induction Therapy & Skip or Delay a Stem Cell Transplant? – SurvivorNet

Cell Therapy in Vascularized Composite Allotransplantation – DocWire News

Posted: January 20, 2022 at 2:20 am

This article was originally published here

Biomed J. 2022 Jan 15:S2319-4170(22)00005-1. doi: 10.1016/j.bj.2022.01.005. Online ahead of print.

ABSTRACT

Allograft rejection is one of the obstacles in achieving a successful vascularized composite allotransplantation. Treatments of graft rejection with lifelong immunosuppression subject the recipients to a lifelong risk of cancer development and opportunistic infections. Cell therapy has recently emerged as a promising strategy to modulate the immune system, minimize immunosuppressant drug dosages, and induce allograft tolerance. In this review, the recent works regarding the use of cell therapy to improve allograft outcomes are discussed. The current data supports the safety of cell therapy. The suitable type of cell therapy in allotransplantation is clinically dependent. Bone marrow cell therapy is more suitable for the induction phase, while other cell therapies are more feasible in either the induction or maintenance phase, or for salvage of allograft rejection. Immune cell therapy focuses on modulating the immune response, whereas stem cells may have an additional role in promoting structural regenerations, such as nerve regeneration. Source, frequency, dosage, and route of cell therapy delivery are also dependent on the specific need in the clinical setting.

PMID:35042019 | DOI:10.1016/j.bj.2022.01.005

More here:
Cell Therapy in Vascularized Composite Allotransplantation - DocWire News

Posted in Stem Cell Therapy | Comments Off on Cell Therapy in Vascularized Composite Allotransplantation – DocWire News

Nowakowski Considers CD19 Therapy in Transplant-Ineligible DLBCL – Targeted Oncology

Posted: January 20, 2022 at 2:20 am

During a Targeted Oncology live event, Grzegorz S. Nowakowski, MD, discussed the case of a patient treated with tafasitamab plus lenalidomide in the second line for diffuse B-cell lymphoma.

Targeted OncologyTM: What are the options for second-line therapy in this patient with DLBCL?

NOWAKOWSKI: The current NCCN [National Comprehensive Cancer Network] guidelines [for patients who are not candidates for transplant] have gemcitabine [Gemzar] plus oxaliplatin [Eloxatin] plus or minus rituximab as a preferred regimen.1

Polatuzumab vedotin [Polivy] plus bendamustine [Treanda] plus rituximab is also included in the NCCN guidelines. Tafasitamab [Monjuvi] plus lenalidomide [Revlimid], which is another option, is FDA approved for second-line therapy and beyond. A lot of us in the field, in patients who are not willing to go for more intensive regimens [such as] transplant or CAR [chimeric antigen receptor] T-cell therapy, are looking more into these chemotherapy combinations, particularly if the patient progresses after chemotherapy. The idea is its going to be a different mode of action. CAR T-cell therapy is [used in the third-line setting] as of now. Again, this may change in the future.

What is the rationale behind the patient receiving this combination?

The FDA granted accelerated approval for the combination of tafasitamab and lenalidomide for relapsed or refractory DLBCL based on [results from] the L-MIND study [NCT02399085].2

Tafasitamab has a cool concept where the antibody cells target CD19, just as in CAR T-cell therapy and loncastuximab tesirine [Zynlonta], which is another recently approved antibody. There were initial developments before studying [CD19] where we felt it could be a good target, but some antibodies didnt work so well. Now there is this renaissance of interest in CD19-targeting agents such as CAR T-cell therapy, tafasitamab, and loncastuximab.

The [tafasitamab] antibody is engineered to have this enhanced Fc function that increases ADCC [antibody-dependent cellular cytotoxicity], ADCP [antibody-dependent cellular phagocytosis], and cell death. It causes direct cell death because CD19 is important in B-cell receptor signaling and not only in the immune system, but it gives some antisignaling properties as well.3

Lenalidomide has the properties of immune activation and microenvironment function and there are dozens of papers postulating many mechanisms of action for lenalidomide. Its very pleiotropic, but it does immune activation, and we know from R2 [lenalidomide plus rituximab] and other antibody combinations that it tends to synergize with the antibodies very well. This preclinical idea led to the development of the combination of this naked antibody and lenalidomide in patients with relapsed or refractory DLBCL.

Which trial data supported the approval of tafasitamab/lenalidomide?

L-MIND was a single-arm, phase 2 study [that enrolled patients who had] 1 to 3 prior regimens and who were either relapsing after transplant or were not eligible for transplant. The primary refractory patients were to be excluded, but because of changing definitions, they accrued, to some degree, to the study, and had pretty good results anyway.4

Tafasitamab is an infusion, just like other antibodies. Its given on days 1, 8, 15, and 22, for 1 to 3 cycles. In cycles 4 to 12, it is given every 2 weeks. Lenalidomide is given at 25 mg daily on days 1 to 25, [just as] in multiple myeloma. This is a different dose [from the R2 regimen], which is 20 mg, but the 25 mg was well tolerated, and this was based on the initial [pilot study]. After 12 cycles of therapy, patients received tafasitamab until disease progression.3,4

Frequently [we are asked] why we would plan on continuing forever. I was involved in the design of the study, and the salvage options for patients were quite limited for those who were not transplant eligible and some of the investigators asked why we would want to stop if it is working. We gave investigators discretion to [decide] whether the patient was benefiting from the treatment and to continue until disease progression. The primary end point of the study was overall response rate [ORR], which has frequently been the most reliable end point for the activity of the combination in this setting because it tends to have less bias in patient selection. The secondary end points were PFS [progression-free survival], duration of response [DOR], overall survival [OS], and so forth.4,5

There were some lenalidomide dose reduction studies where patients were given doses of 25 mg down to 5 mg using step reductions.5

This was a study of the [safety] population, and 81 patients were accrued overall. The median age was 72. The IPI risk score, Ann Arbor stage, and LDH results were typical for refractory DLBCL. Patients with primary refractory disease were supposed to be excluded, but 19 of 81 patients had it and 44 of 81 patients were refractory to prior therapies. Relatively few patients had a prior stem cell transplant and the majority were not eligible for it due to comorbidities, unwillingness to do so, or not responding to salvage therapy. [Not responding] to previous therapies was a major reason [for not getting a transplant].5

How did patients do on the L-MIND trial?

The ORR for this combination was quite high at greater than 60%, which is comparable with what we see in CAR T-cell therapy or intensive chemotherapy. So this was quite significant and impressive at the time the [results were] published. The CR [complete response] rate was even more impressive at 43%. Again, this was in patients who were relapsed or refractory, not transplant eligible, or those relapsing after transplant, so a 43% CR rate is high.5,6

As clinicians, we care about the DOR, too. So if you are a regulator, say at the FDA, you only worry about response rates because its less about patient selection, but clinicians like responses to be durable. The median PFS was 12.1 months.5 The median PFS doesnt fully reflect the activity of this regimen because it plateaus just after the median. CAR T-cell therapy data look very similar, too. For a relatively well-tolerated combination, these were very impressive results at the time of presentation. The median OS was not reached and, as with the PFS results, the OS also plateaued. So these were very impressive results in terms of DOR.

The patients in CR were primarily driving this benefit, but even the patients in PR [partial response] had [an approximately] 30% sustained response.6 The treatment was active in the patients treated both with 1 prior or 2 or more prior lines of therapy. Responses, particularly the CR rates, were somewhat higher in the patients who were on second-line treatment. This would be the patients who were not eligible for transplant.

Do you feel comfortable using this regimen in patients with GCB [germinal center B-celllike] subtypes because they were underrepresented in the study?

There was a whole debate about it. We believe that the combination of the antibodies and lenalidomide works well in GCB subtypes as well. It is a little bit different with single agents because the data showed response rates and activity were better in ABC [activated B-cell] or nonGCB subtypes of DLBCL, but in combination, there appeared to be less of a differential by cell of origin.

But in the [forest plot] analysis, both subtypes benefited. There was a trend toward a little bit of a high response rate in patients with the ABC subtype, but overall, the response rate was high in patients with GCB patients as well. I believe it was approximately 45% to 50% in both subtypes.

What about the R2 regimen? Do you prefer not to use it in GCB subtypes?

Yes, I prefer not to use it in GCB subtypes. [Results of] the ECOG-ACRIN E1412 study [NCT01856192] were recently published in the Journal of Clinical Oncology and I was a PI [principal investigator] in it.7 This study was looking at all-comers, so it was the only randomized frontline phase 2 study, where lenalidomide was added to R-CHOP. This one was cell-of-origin agnostic, so they could have the GCB or ABC subtype. There was [approximately] a 12% difference in PFS in this study and a favorable hazard ratio.

Another study, the ROBUST study [NCT02285062], was focused on patients with the ABC subtype.8 It didnt show a difference using different lenalidomide scheduled doses, though there were other patient selection issues in the study. As a single agent, lenalidomide is more active in the ABC subtype and I use it myself in clinical practice more in ABC or nonGCB subtypes. In combination with the antibodies, or even chemotherapy, this may not be necessarily true. Because most of these patients are already exposed to rituximab, I think based on the R2 study [results], they didnt see much of a differential based on cell of origin, which is a little bit disappointing, because we were hoping we could [use it to] select the high responders, but that didnt pan out. REMARC [NCT01122472] was a study done by a French group that used lenalidomide maintenance after R-CHOP but didnt track the cell of origin.

In fact, the GCB subtype tended to benefit more, and an idea was that maybe some microenvironment influences played a role. In my clinical practice, in nonGCB subtypes, I use a single agent, but for combination of the antibodies, the activity seems to be agnostic to cell of origin.

How does an anti-CD19 antibody downregulate the CD19 receptor?

There is limited information, but they did a study looking at the CD19 expression after tafasitamab exposure in chronic lymphocytic leukemia and [there was no impact] and in DLBCL as well. The CD19 expression is just a part of the story because you worry that a part of the CD19 molecule could be mutated and then the CAR T-cell agents would not bind or that part of the molecule could be missed because of alternative splicing or losing one of the exons because of the evolutionary pressure of the treatment. We did whole exome and RNA sequencing and saw no abnormalities within the CD19 cells. It appears to be expressed after tafasitamab exposure, and there are no point mutations, exon deletions, or other changes that would affect the integrity of CD19, to the best of our knowledge.

Of course, the best data would come from clinical evidence if we note that CAR T-cell therapy is working. In this study, only 1 patient proceeded with CAR T-cell therapy and had good clinical benefit and was in remission last time I saw the data. So it appears that in anecdotal experiences CAR T-cell therapy will still work in those patients.

The opposite is true, too. There is a huge interest now in this combination and [whether] it will be active in post CAR T-cell relapses. Lenalidomide as a single agent is frequently used in this setting. How active will this combination be in postCAR T-cell relapse? We know that lenalidomide is active. A lot of patients with CAR T-cell relapses will still have CD19, so we believe that is also an option, but more data will be needed.

Do patients tolerate the 25-mg lenalidomide dose in combination with tafasitamab, or is the dose modified often?

[Approximately] 30% of patients will have to drop to 20 mg, particularly with subsequent cycles. The nice thing for lenalidomide is that you can use the growth factor support because it is primarily neutropenia that causes some of the dose reductions. Studies are different from real life, so in the real world we always have some patients who are already cytopenic from the previous therapy. I usually support them with a growth factor, and sometimes I start my patients at 20 mg. The dosing intensity of lenalidomide seems to be important, though.

I wouldnt very liberally decrease it because there appears to be some dose relation to the response, at least as a single agent in a refractory setting in DLBCL in contrast to follicular [lymphoma], but somewhere from 15 mg to 20 mg is the golden spot for response.

The 25 mg was used in those studies as a single agent, so, about one-third of patients did require dose reductions. If you use this combination, you follow the lenalidomide package inserts, and if you need to reduce because of creatinine clearance, you reduce the lenalidomide or if you see significant neutropenia despite the growth factor used, then you can reduce on a subsequent cycle to 20 mg, or interrupt and reduce to 20 mg.

Does patient preference weigh into the decision to choose finite therapy vs therapy until progression of disease in the second-line setting?

Yes, it comes down to the patients preference. I dont practice in the community, so I dont have more experience with this. We have this policy at Mayo Clinic that [any clinician] from around the world can call us at any time for advice about their patients. So, routinely, we are getting quite a few phone calls from those who are responsible for patients with lymphoma, or for any other disease type from outside, and practitioners call asking what to do.

I am always surprised by how many patients do not want to proceed with CAR T-cell therapy or stem cells or even clinical trials, which we often have here, because of the preference of being near the local center. Travel is not always possible and some patients want to stay where they are, which is a very reasonable option.

Are there trials comparing this with transplant or something lenalidomide alone?

We did 2 things to differentiate this from lenalidomide alone. A study called RE-MIND [NCT04150328] with close matching of the patients with real-world data showed that the combination was definitely much more active than lenalidomide alone. [We knew this] but wanted to double-check in a very close-matched cohort. A confirmatory study for this is [the frontMIND study (NCT04824092), which is a frontline study that compares] R-CHOP as standard therapy vs R2-CHOP plus tafasitamab.

I am the principal investigator globally for this study, and one of the reasons why we designed it this way was there was some activity already from randomized phase 2 studies using lenalidomide. It was safe and effective and also the doublet was already approved, so it was logical to move it forward.

However, the biggest [issue we had when] presenting this concept to some regulatory authorities was that we were a little bit naive in the past, thinking that adding 1 drug at a time is going to move the bar a whole lot. R-CHOP already has 5 different compounds, so I think the sixth one probably is not going to move the bar a whole lot. There are some studies that failed, I think, 1 drug at a time. So the ambitious plan here is to add a doublet. But the study is designed to capture very high-risk patients, [meaning] IPI 3 and above. Its looking at the highest-risk population and is adding doublet on top of R-CHOP. There are some study centers in the United States that are in the process of either opening or even have it open currently.

Could tafasitamab/lenalidomide be moved to the first-line setting with more targeted agents as chemotherapies are eliminated?

Yes. There is a pilot study led by my colleague Dr [Jason] Westin at [The University of Texas MD Anderson Cancer Center]. He is basically pioneering the so-called smart-start, or smart-stop now, where he is adding exactly this combination to R-CHOP. The question is: Can he strip some of the chemotherapy agents [such as anthracyclines]?

[The patient] tried to shorten and then to remove different cytotoxic drugs with the idea that maybe over time he can develop a chemotherapy-free regimen. [Results of] the initial pilot study have shown this combination plus ibrutinib [Imbruvica] is producing high response rates. He still added chemotherapy later because he was worried that he may miss the possibility of curing the patient, but after initial feasibility, he is slowly stripping chemotherapy. We may get there one day.

What are the similarities and differences of loncastuximab tesirine and tafasitamab?

I think cross-study comparisons are usually difficult. I am very cautious always when comparing different study results because the patient population is not always the same. I happen to be involved with the FDA in different reviews and I do believe that the response rate is what tends to reflect the most activity and is less dependent on patient selection, though not completely.

The ORR of loncastuximab is [approximately] 50% or very close to that. The DOR appears to be a little bit shorter, but this could be due to patient selection, so it looks very encouraging. It has a little bit of a different adverse event [AE] profile. At this point it doesnt have as strong a follow-up as this study, so we dont know if the same very encouraging plateaus in responding patients will be seen with it.

Maybe its going to happen, but it is more of a traditional cytotoxic therapy that is directed like polatuzumab. It works more on the immune microenvironment in immune activation. There is this renaissance of CD19 targeting and for CAR T-cell therapies, all the approved products target CD19, and now loncastuximab and tafasitamab.

I usually tell the industry to not develop any more agents targeting CD19. We have enough. There are some other good targets, too. Some of the CAR T-cell therapies are targeting different molecules on the surface.

How many of these patients on the L-MIND trial stopped therapy early? What is the safety profile of combination lenalidomide and tafasitamab?

The primary reason for stopping therapy early was disease progression because some patients just didnt respond. The toxicities were primarily hematologic, which is consistent with what you would see with lenalidomide. Nonhematologic AEs [included] fatigue and diarrhea, but nothing striking or unusual. Discontinuation of combination [therapy due to] AEs was seen in 12% of the patients [n = 10/81].5

A comparison of the AEs of combination therapy vs monotherapy showed the hematologic and other toxicities were driven by lenalidomide. Tafasitamab alone had [an approximate] 27% ORR and when combined with lenalidomide the response rate doubles, so theres a true synergy between those drugs.

The monotherapies are quite well tolerated. Some patients can develop neutropenia, as was seen in the monotherapy trials, but overall the toxicity is minimal for the antibody alone.

What is the rapidity of the response for this regimen? Who wouldnt be eligible for it?

The first evaluation was done after 2 cycles of therapy, so within 8 weeks the response was right there. The response is quite brisk. If I had any concern about putting [a patient] on lenalidomide, it would be for reasons such as it can cause some rashes as seen previously with lenalidomide combinations, so with previous hypersensitivity, I probably would not [use it].

If patients have very rapidly progressive symptoms, I may stabilize them with radiation or some other treatment first, maybe hydroxysteroids, rituximab, or something such as that just to remove the disease burden before I start this combination. I expected that the responses would be dipping over time, but the responses were brisk and happened after 2 cycles of therapy.

REFERENCES

Here is the original post:
Nowakowski Considers CD19 Therapy in Transplant-Ineligible DLBCL - Targeted Oncology

Posted in Stem Cell Therapy | Comments Off on Nowakowski Considers CD19 Therapy in Transplant-Ineligible DLBCL – Targeted Oncology

Bone Therapeutics provides fourth quarter 2021 business update and 2022 outlook – GlobeNewswire

Posted: January 20, 2022 at 2:20 am

REGULATED INFORMATION

Recruitment for ALLOB tibial facture Phase IIb study ongoing and on schedule to release topline data in Q1 2023, despite COVID slowdown

New scientific advisory board appointments to bolster the further development of the next generation iMSCg platform

Discussions for ALLOB global partnership still ongoing. Completion of potential global partnership delayed and now anticipated in Q1 2022

Financial position strengthened following equity fundraising in Q4 2021 with runway expected into Q3 2022

Gosselies, Belgium, 19 January 2022, 7am CET BONE THERAPEUTICS (Euronext Brussels and Paris: BOTHE), the cell therapy company addressing unmet medical needs in orthopedics and other diseases, announces today a business update for the fourth quarter, ending 31 December 2021 as well as a business outlook for 2022.

Bone Therapeutics activity in Q4 2021 has resulted in a broadened pipeline and stronger therapeutic potential for the company. The pandemic continues to be a concern across the cell and gene therapy, biopharma and healthcare industries. Bone Therapeutics has, however, put measures in place to reduce the impact of the pandemic on its clinical development., said Miguel Forte, Chief Executive Officer, Bone Therapeutics. In addition, the deal for the global rights for the allogeneic osteoblastic cell therapy product ALLOB, when and if completed, will allow Bone Therapeutics, together with its partners, to ensure the development of ALLOB towards commercialization, while at the same time continue to explore options to expand the iMSCg platform. This includes the development of a next generation of genetically engineered mesenchymal stromal cells and the use of highly scalable and versatile cell sources such as induced pluripotent stem cells. Bone Therapeutics has expanded its Scientific Advisory Board purely for this purpose.

Operational highlights

Corporate highlights

Financial highlights (1)

Outlook for 2022

Financial Calendar 2022

The financial calendar is communicated on an indicative basis and may be subject to change.

(1) Unaudited number

About Bone Therapeutics

Bone Therapeutics is a leading biotech company focused on the development of innovative products to address high unmet needs in orthopedics and other diseases. The Company has a diversified portfolio of cell therapies at different stages ranging from pre-clinical programs in immunomodulation to mid stage clinical development for orthopedic conditions, targeting markets with large unmet medical needs and limited innovation.

Bone Therapeutics core technology is based on its cutting-edge allogeneic cell and gene therapy platform with differentiated bone marrow sourced Mesenchymal Stromal Cells (MSCs) which can be stored at the point of use in the hospital. Currently in pre-clinical development, BT-20, the most recent product candidate from this technology, targets inflammatory conditions, while the leading investigational medicinal product, ALLOB, represents a unique, proprietary approach to bone regeneration, which turns undifferentiated stromal cells from healthy donors into bone-forming cells. These cells are produced via the Bone Therapeutics scalable manufacturing process. Following the CTA approval by regulatory authorities in Europe, the Company has initiated patient recruitment for the Phase IIb clinical trial with ALLOB in patients with difficult tibial fractures, using its optimized production process. ALLOB continues to be evaluated for other orthopedic indications including spinal fusion, osteotomy, maxillofacial and dental.

Bone Therapeutics cell therapy products are manufactured to the highest GMP (Good Manufacturing Practices) standards and are protected by a broad IP (Intellectual Property) portfolio covering ten patent families as well as knowhow. The Company is based in the BioPark in Gosselies, Belgium. Further information is available at http://www.bonetherapeutics.com.

For further information, please contact:

Bone Therapeutics SAMiguel Forte, MD, PhD, Chief Executive OfficerLieve Creten, Chief Financial Officer ad interimTel: +32 (0)71 12 10 00investorrelations@bonetherapeutics.com

For Belgian Media and Investor Enquiries:BepublicBert BouserieTel: +32 (0)488 40 44 77bert.bouserie@bepublicgroup.be

International Media Enquiries:Image Box CommunicationsNeil Hunter / Michelle BoxallTel: +44 (0)20 8943 4685neil.hunter@ibcomms.agency / michelle@ibcomms.agency

For French Media and Investor Enquiries:NewCap Investor Relations & Financial CommunicationsPierre Laurent, Louis-Victor Delouvrier and Arthur RouillTel: +33 (0)1 44 71 94 94bone@newcap.eu

Certain statements, beliefs and opinions in this press release are forward-looking, which reflect the Company or, as appropriate, the Company directors current expectations and projections about future events. By their nature, forward-looking statements involve a number of risks, uncertainties and assumptions that could cause actual results or events to differ materially from those expressed or implied by the forward-looking statements. These risks, uncertainties and assumptions could adversely affect the outcome and financial effects of the plans and events described herein. A multitude of factors including, but not limited to, changes in demand, competition and technology, can cause actual events, performance or results to differ significantly from any anticipated development. Forward looking statements contained in this press release regarding past trends or activities should not be taken as a representation that such trends or activities will continue in the future. As a result, the Company expressly disclaims any obligation or undertaking to release any update or revisions to any forward-looking statements in this press release as a result of any change in expectations or any change in events, conditions, assumptions or circumstances on which these forward-looking statements are based. Neither the Company nor its advisers or representatives nor any of its subsidiary undertakings or any such persons officers or employees guarantees that the assumptions underlying such forward-looking statements are free from errors nor does either accept any responsibility for the future accuracy of the forward-looking statements contained in this press release or the actual occurrence of the forecasted developments. You should not place undue reliance on forward-looking statements, which speak only as of the date of this press release.

More:
Bone Therapeutics provides fourth quarter 2021 business update and 2022 outlook - GlobeNewswire

Posted in Stem Cell Therapy | Comments Off on Bone Therapeutics provides fourth quarter 2021 business update and 2022 outlook – GlobeNewswire

Omeros Confirms Submission of Response to FDA Regarding the BLA for Narsoplimab in the Treatment of HSCT-TMA – Business Wire

Posted: January 20, 2022 at 2:20 am

SEATTLE--(BUSINESS WIRE)--Omeros Corporation (Nasdaq: OMER) today confirmed that earlier this month the company submitted to the U.S. Food and Drug Administration (FDA) its response to the Agencys Complete Response Letter (CRL) for narsoplimab in the treatment of hematopoietic stem cell transplant-associated thrombotic microangiopathy (HSCT-TMA). The response comprises a comprehensive briefing package drafted in close collaboration with external clinical, regulatory and legal experts that addresses in detail the points raised by FDA in its CRL for narsoplimab. Omeros concurrently requested a Type A meeting with FDA to resolve any outstanding items.

Narsoplimab is the first drug candidate submitted to FDA for approval in HSCT-TMA. It has Breakthrough Therapy and Orphan designations in both HSCT-TMA and IgA nephropathy.

About Omeros Corporation

Omeros is an innovative biopharmaceutical company committed to discovering, developing and commercializing small-molecule and protein therapeutics for large-market and orphan indications targeting inflammation, immunologic diseases (e.g., complement-mediated diseases) and cancers. Omeros lead MASP-2 inhibitor narsoplimab targets the lectin pathway of complement and is the subject of a biologics license application pending before FDA for the treatment of hematopoietic stem cell transplant-associated thrombotic microangiopathy. Narsoplimab is also in multiple late-stage clinical development programs focused on other complement-mediated disorders, including IgA nephropathy, atypical hemolytic uremic syndrome and COVID-19. OMS906, Omeros inhibitor of MASP-3, the key activator of the alternative pathway of complement, is in a Phase 1 clinical trial. For more information about Omeros and its programs, visit http://www.omeros.com.

Forward-Looking Statements

This press release contains forward-looking statements within the meaning of Section 27A of the Securities Act of 1933 and Section 21E of the Securities Exchange Act of 1934, which are subject to the safe harbor created by those sections for such statements. All statements other than statements of historical fact are forward-looking statements, which are often indicated by terms such as anticipate, believe, could, estimate, expect, goal, intend, likely, look forward to, may, objective, plan, potential, predict, project, should, slate, target, will, would and similar expressions and variations thereof. Forward-looking statements, including expectations with regard to interactions and communications with FDA and Omeros pursuit of regulatory approval for narsoplimab in HSCT-TMA, are based on managements beliefs and assumptions and on information available to management only as of the date of this press release. Omeros actual results could differ materially from those anticipated in these forward-looking statements for many reasons, including, without limitation, risks associated with product commercialization and commercial operations, regulatory processes and oversight, and the risks, uncertainties and other factors described under the heading Risk Factors in the companys Annual Report on Form 10-K filed with the Securities and Exchange Commission on March 1, 2021. Given these risks, uncertainties and other factors, you should not place undue reliance on these forward-looking statements, and the company assumes no obligation to update these forward-looking statements, whether as a result of new information, future events or otherwise, except as required by applicable law.

See original here:
Omeros Confirms Submission of Response to FDA Regarding the BLA for Narsoplimab in the Treatment of HSCT-TMA - Business Wire

Posted in Stem Cell Therapy | Comments Off on Omeros Confirms Submission of Response to FDA Regarding the BLA for Narsoplimab in the Treatment of HSCT-TMA – Business Wire

City of Hope and CytoImmune announce study demonstrating novel off-the-shelf chimeric antigen receptor (CAR) natural killer (NK) cell-based therapy…

Posted: January 20, 2022 at 2:20 am

News and research before you hear about it on CNBC and others. Claim your 1-week free trial to StreetInsider Premium here.

DUARTE, Calif. & TOA BAJA, Puerto Rico--(BUSINESS WIRE)--City of Hope, a world-renowned cancer research and treatment organization, and CytoImmune Therapeutics, a clinical-stage immunotherapy company that is developing a novel class of natural killer (NK) cell-based cancer therapies, today announced a study published in the high-impact journal Gastroenterology that demonstrates off-the-shelf anti-prostate stem cell antigen (PSCA) chimeric antigen receptor (CAR) NK cells significantly suppressed pancreatic cancer in vitro and in vivo using a method known as freeze-thaw.

This press release features multimedia. View the full release here: https://www.businesswire.com/news/home/20220118005366/en/

The therapy PSCA CAR_s15 NK cells, also known as CYTO NK-203 persisted more than 90 days after infusion and significantly prolonged the survival of mice with pancreatic cancer, showing that the freeze-thaw method works. For the study, PSCA CAR_s15 NK cells were produced and then frozen. The cells were then thawed and used in preclinical studies at City of Hope.

Our patients need additional ways to attack their pancreatic cancer. The work presented by City of Hopes team is distinctive and promising for two reasons: First of all, it is based on a precision medicine approach that is a special target in the patient's pancreatic cancer PSCA. Secondly, it is an immunologic approach, using human natural killer cells, which are specifically engineered to attack the patient's cancer. These findings should be accelerated to a clinical trial as rapidly as possible, said Daniel D. Von Hoff, M.D., a distinguished professor in the Molecular Medicine Division of the Translational Genomics Research Institute (TGen), an affiliate of City of Hope. He also is senior consultant-clinical investigator at City of Hope and is one of the nations leading authorities on the treatment and care of pancreatic cancer patients.

City of Hope is committed to finding more effective and innovative treatments for difficult-to-treat solid cancers, and pancreatic cancer is clearly one of them, said Saul Priceman, Ph.D., assistant professor in the Department of Hematology & Hematopoietic Cell Transplantation at City of Hope and a study author. These new PSCA CAR_s15 NK cell preclinical studies provide tremendous support for the anticipated upcoming clinical trials to evaluate efficacy and safety of this novel CAR-engineered NK cell therapy in patients with pancreatic cancer, which is a promising expansion of our existing clinical programs that target PSCA in solid cancers using CAR-engineered T cell therapy.

Pancreatic cancer is the third leading cause of cancer-related death in the United States with a five-year survival rate of approximately 10%. The cancer is typically detected when it is at a late stage and incurable. Chemotherapy or other therapies provide modest benefit. Therefore, the development of new therapies for pancreatic cancer is crucial. The therapy can also be used for other PSCA+ cancers, such as stomach and prostate.

NK cell technology works by using natural killer cells from a patient or donor. NK cells are then engineered so they express a receptor a CAR that is specific for a protein expressed by cancerous cells, along with the secretion of IL-15, which sustains the survival of the NK cells.

Christina Coughlin, M.D., CEO of CytoImmune Therapeutics, said, "We are excited to share this data on our CAR NK candidate for pancreatic cancer. This foundational data supports robust anti-tumor activity with CYTO NK-203, making us confident our innovative and off-the-shelf NK cell therapy approach has the potential to deliver more accessible, safe and effective cell-based treatment options to cancer patients. We are encouraged by these findings and look forward to continuing our work with City of Hope in order to move this initiative to the clinic.

This immunotherapy is revolutionizing the treatment of some blood cancers; however, its use in the treatment of solid tumors has been limited, in part because most of the proteins currently used to target CAR cells to solid tumors are present in low levels on other normal tissues, leading to toxic side effects.

Based on research by Michael Caligiuri, M.D., president of City of Hope National Medical Center and the Deana and Steve Campbell Physician-in-Chief Distinguished Chair, and Jianhua Yu, Ph.D., professor and director of the Natural Killer Cell Biology Research Program, who have nearly 55 years of collective laboratory investigation of NK cells, CytoImmune is developing an NK cell platform designed to overcome the limitations and challenges of current technologies for engineering NK cells. The platform is designed to generate an abundant supply of CAR NK cells from a single umbilical cord donor, engineered with the CAR for effective recognition of tumor targets, and secreting IL-15 to improve the persistence of CAR NK cells for sustained activity in the body. The process enables scientists to freeze, transport and store engineered CAR NK cells for off-the-shelf use for the treatment of cancer.

The study titled Off-the-shelf PSCA-directed chimeric antigen receptor natural killer cell therapy to treat pancreatic cancer can be found here.

About City of Hope

City of Hope is an independent biomedical research and treatment center for cancer, diabetes and other life-threatening diseases. Founded in 1913, City of Hope is a leader in bone marrow transplantation and immunotherapy such as CAR T cell therapy. City of Hopes translational research and personalized treatment protocols advance care throughout the world. Human synthetic insulin, monoclonal antibodies and numerous breakthrough cancer drugs are based on technology developed at the institution. A National Cancer Institute-designated comprehensive cancer center and a founding member of the National Comprehensive Cancer Network, City of Hope is ranked among the nations Best Hospitals in cancer by U.S. News & World Report. Its main campus is located near Los Angeles, with additional locations throughout Southern California and in Arizona. Translational Genomics Research Institute (TGen) became a part of City of Hope in 2016. AccessHopeTM, a subsidiary launched in 2019, serves employers and their health care partners by providing access to NCI-designated cancer center expertise. For more information about City of Hope, follow us on Facebook, Twitter, YouTube or Instagram.

About CYTO NK 203

CYTO NK-203 is an off-the-shelf allogeneic CAR NK cell therapy derived from umbilical cord blood, expressing a CAR against prostate stem cell antigen and soluble IL-15 and engineered with proprietary features designed to improve the safety and efficacy of NK cells as a potential therapy.

About CytoImmune Therapeutics Inc.

Founded in 2017, CytoImmune Therapeutics is a clinical-stage biotechnology company, focused on developing an innovative and differentiated pipeline of NK cell therapies, using proprietary, robust and well characterized NK cell expansion and engineering technologies pioneered by Michael Caligiuri, M.D., and Jianhua Yu, Ph.D. The pipeline includes cytokine induced NK (CI-NK) for lung cancer, FLT3 CAR-NK for acute myeloid leukemia, PSCA CAR-NK cells for solid tumors and GPRC5D BiKE secreting BCMA CAR-NK cells for multiple myeloma. CytoImmunes lead product, CYTO-102 (CI-NK) cell therapy, aims to enter the clinic in combination with atezolizumab (anti-PD-L1 monoclonal antibody) for nonsmall cell lung cancer in 2022.

View source version on businesswire.com: https://www.businesswire.com/news/home/20220118005366/en/

City of HopeLetisia Marquezlemarquez@coh.org626-476-7593

CytoImmuneWilliam Roselliniwill@cytoimmune.com

Source: City of Hope

See the rest here:
City of Hope and CytoImmune announce study demonstrating novel off-the-shelf chimeric antigen receptor (CAR) natural killer (NK) cell-based therapy...

Posted in Stem Cell Therapy | Comments Off on City of Hope and CytoImmune announce study demonstrating novel off-the-shelf chimeric antigen receptor (CAR) natural killer (NK) cell-based therapy…

Cynata Therapeutics scoops bumper R&D incentive – The West Australian

Posted: January 20, 2022 at 2:20 am

Clinical-stage biotechnology company, Cynata Therapeutics has lopped over $800,000 off its tax bill after the companys cell therapeutic work was deemed eligible for the Federal Governments research and development tax refund scheme. The company says the savings will be ploughed into its expansive series of clinical trials which include osteoarthritis, respiratory failure, diabetic foot ulcers and its imminent graft-versus-host disease trials.

The refund covers work completed over the 2020/2021 financial year and boosts Cynatas already healthy cash position, reported at $23.9m in recent financial reports.

The rebate is courtesy of the Australian Governments Research and Development Tax Incentive. The scheme has served as the Federal Governments primary means of supporting domestic innovation and has helped drive a suite of ground-breaking products and services since its introduction more than a decade ago. The system provides an opportunity for eligible local and foreign companies undertaking R&D activities in Australia to reduce their tax bill by providing a rebate on activities that meet the criteria.

The Victorian-based company has been on the receiving-end of a number of bumper financial plays of late, including $5 million from specialised human cell manufacturing heavyweight, Fujifilm Cellular Dynamics under a partnership agreement inked by the duo in October.

That deal will see Fujifilm provide clinical and commercial manufacturing services for Cynatas Cymerus therapeutic mesenchymal stem cell products.

Cynata says its Cymerus products offer superior benefits to alternative products on the market. According to the company, Cymerus, unlike its competitive products, uses a single donor to produce cell therapy products at a commercial scale whilst alternative stem cell treatments require multiple donors.

The companys recent spate of activity also saw it regain commercialisation rights to its CYP-001 as a treatment for graft versus host disease. The illness typically rears its head after bone marrow transplants and is typified by the donors immune cells attacking the recipient of the transplant.

With a healthy financial injection back into in the companys coffers, Cynata can focus its efforts on further developing its comprehensive and cutting-edge product pipeline in the lucrative clinical-stage biotechnology space.

Is your ASX-listed company doing something interesting? Contact: matt.birney@wanews.com.au

Read more:
Cynata Therapeutics scoops bumper R&D incentive - The West Australian

Posted in Stem Cell Therapy | Comments Off on Cynata Therapeutics scoops bumper R&D incentive – The West Australian

The role of gel wound dressings loaded with stem cells in the treatment of diabetic foot ulcers – DocWire News

Posted: January 20, 2022 at 2:20 am

This article was originally published here

Am J Transl Res. 2021 Dec 15;13(12):13261-13272. eCollection 2021.

ABSTRACT

Diabetic foot ulcers (DFUs) are a serious complication of diabetes and the main cause of nontraumatic lower limb amputations, resulting in a serious economic burden on society. The main causes of DFUs include peripheral neuropathy, foot deformity, chronic inflammation, and peripheral artery disease. There are many clinical approaches for the treatment of DFUs, but they are all aimed at addressing a single aetiological factor. Stem cells (SCs), which express many cytokines and a variety of nerve growth factors and modulate immunological function in the wound, may accelerate DFU healing by promoting angiogenesis, cell proliferation, and nerve growth and regulating the inflammatory response. However, the survival time of SCs without scaffold support in the wound is short. Multifunctional gel wound dressings play a critical role in skin wound healing due to their ability to maintain SC survival for a long time, provide moisture and prevent electrolyte and water loss in DFUs. Among the many methods for clinical treatment of DFUs, the most successful one is therapy with gel dressings loaded with SCs. To accelerate DFU healing, gel wound dressings loaded with SCs are needed to promote the survival and migration of SCs and increase wound contraction. This review summarizes the research advancements regarding multifunctional gel wound dressings and SCs in the treatment of DFU to demonstrate the effectiveness and safety of this combinational therapeutic strategy.

PMID:35035674 | PMC:PMC8748097

Go here to see the original:
The role of gel wound dressings loaded with stem cells in the treatment of diabetic foot ulcers - DocWire News

Posted in Stem Cell Therapy | Comments Off on The role of gel wound dressings loaded with stem cells in the treatment of diabetic foot ulcers – DocWire News

Cancer stem cells in glioblastoma

Posted: January 20, 2022 at 2:17 am

Genes Dev. 2015 Jun 15; 29(12): 12031217.

1Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA;

2Department of Molecular Medicine, Cleveland Clinic, Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195, USA;

3Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA

1Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA;

3Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA

2Department of Molecular Medicine, Cleveland Clinic, Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195, USA;

3Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA

1Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA;

2Department of Molecular Medicine, Cleveland Clinic, Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195, USA;

3Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA

Tissues with defined cellular hierarchies in development and homeostasis give rise to tumors with cellular hierarchies, suggesting that tumors recapitulate specific tissues and mimic their origins. Glioblastoma (GBM) is the most prevalent and malignant primary brain tumor and contains self-renewing, tumorigenic cancer stem cells (CSCs) that contribute to tumor initiation and therapeutic resistance. As normal stem and progenitor cells participate in tissue development and repair, these developmental programs re-emerge in CSCs to support the development and progressive growth of tumors. Elucidation of the molecular mechanisms that govern CSCs has informed the development of novel targeted therapeutics for GBM and other brain cancers. CSCs are not self-autonomous units; rather, they function within an ecological system, both actively remodeling the microenvironment and receiving critical maintenance cues from their niches. To fulfill the future goal of developing novel therapies to collapse CSC dynamics, drawing parallels to other normal and pathological states that are highly interactive with their microenvironments and that use developmental signaling pathways will be beneficial.

Keywords: brain tumor, cancer stem cell, glioblastoma, glioma, stem cell, tumor-initiating cell

Development is a coordinated summation of the individual cellular dynamics that build an organ, and the programs responsible for this construction are generally preserved in stem cells for organ homeostasis and tissue repair. Tumors are complex systems that recapitulate the complexity of organs or tissues with dynamic regulation and constituent cellular populations during tumor initiation, maintenance, and progression (Hanahan and Weinberg 2011). While many scientists have sought to reduce the complexity of cancer to a one-dimensional processfor example, characterizing cancers solely based on geneticsmost advanced cancers unfortunately remain nearly as lethal since the declaration of the War on Cancer in 1971. Targeted therapeutics offer a transient benefit for some cancer types with driving mutations, but even these tumors will develop resistance to overcome initially effective therapies that poison driving oncogenes.

Glioblastoma (GBM; World Health Organization grade IV glioma) is the most prevalent and lethal primary intrinsic brain tumor (Stupp et al. 2009). Unlike other solid tumor cell types, GBM widely invades the surrounding brain but rarely metastasizes to other organs. While halting steps to fight GBM are being made using targeted therapies (e.g., bevacizumab) or immunotherapies, GBM therapy remains focused on achieving maximal surgical resection followed by concurrent radiation therapy with temozolomide (TMZ; an orally available methylation chemotherapy) and subsequent additional adjuvant TMZ therapy. Conventional treatment offers patients with GBM additional survival time with generally acceptable quality of life, but a cure is never achieved. GBM represents one of the most comprehensively genomically characterized cancer types, leading to recognition of groups of tumors defined by transcription profiles (proneural, neural, classical, and mesenchymal), genetics (mutations of isocitrate dehydrogenase 1 [IDH1]), and epigenetics (CpG island methylator phenotype [CIMP]) (Weller et al. 2013). Long-term survivors are often, but not exclusively, patients with tumors harboring mutations in IDH1, which likely represent a different disease than most GBMs. Beyond IDH1 mutations and a few other biomarkers (deletion of chromosomes 1p and 19q in oligodendrogliomas, methyl guanine methyltransferase [MGMT] promoter methylation, etc.), the accumulated genetic characterization of GBMs has failed to impact clinical practice, suggesting that other discovery paradigms should also be considered.

The brain, like other organs with clearly defined cellular hierarchies in development and homeostasis (e.g., blood, breast, skin, and colon), gives rise to tumors with defined cellular hierarchies, suggesting that cancer replicates ontogeny (Reya et al. 2001). Atop the apex of cellular hierarchies are stem cells, which have been assumed to be rare, quiescent, self-renewing cells, but several highly proliferative organs (e.g., the intestine and skin) contain at least two pools of stem cells: one quiescent, and the other highly proliferative (Barker et al. 2010). Stem cells generate transient amplifying cells, which in turn create lineage-restricted progeny that are eventually fated to become the terminally differentiated effector cells.

Neural stem and progenitor cell (NSPC) pools vary in location and control during development, suggesting that different cellular hierarchies may be co-opted by brain tumors (Gibson et al. 2010; Lottaz et al. 2010). Informed by techniques used to enrich and characterize NSPCs, several groups in parallel demonstrated that gliomas and other primary brain tumors contain self-renewing, tumorigenic cells (Ignatova et al. 2002; Hemmati et al. 2003; Singh et al. 2003, 2004; Galli et al. 2004). The nomenclature for these cells has been controversial (as discussed below), with the dominant choice being cancer stem cells (CSCs) or tumor-initiating cells (importantly, these terms are not identical, as a CSC designation is more restrictive but also more informative) (). Unlike normal brain organizationwhere the generation of differentiated progeny is stage-specific (neurons and then glia during development) and derived from rapidly dividing progenitor cells and quiescent, multipotent stem cells that persist into adulthood and become activated upon differentiation (Rowitch and Kriegstein 2010)these populations have yet to be delineated in brain tumors. The ability to distinguish between self-renewing cells with stem and progenitor cell cycle properties and transcriptional signatures is likely to provide clarity with respect to nomenclature and the functional interplay between cells at the apex of the hierarchy. The challenges distinguishing CSCs from their progeny are derived, in part, from the limited recognition of points of relative stability (attractor states) in the landscape of cellular identity that define the stem cell state and transitions into (dedifferentiation) and out of (differentiation) a stable state (Chang et al. 2008). Much like the Heisenberg uncertainty principle in physics, our ability to observe the state of a cell is limited by our act of measurement. The presence of subatomic particles is confirmed in retrospect; similarly, the functional definition of both normal and neoplastic stem cells requires retrospective confirmation. The ability to prospectively distinguish glioma stem cells, which reside at the apex of tumor hierarchies, from their differentiated progeny remains challenging; however, stem cell biology faces a similar difficulty with normal stem cell identification. Of note, the CSC hypothesis does not claim a stem cell as the cell of origin for cancers, suggesting that CSCs do not need to adhere to all of the observed features of normal stem cells.

Definitions and functional characteristics of CSCs, tumor-initiating cells, and tumor-propagating cells

In the following sections, we provide an update on intrinsic and extrinsic regulators of the CSC state in GBM and discuss how the integration of genetics, epigenetics, and metabolism has shaped our understanding of how CSCs function to drive GBM growth. We also highlight future opportunities to further understand the complexity of CSC regulation through interaction with other cells (including immune cells) and how the translation of CSC-based therapies needs to take into account the cellular dynamics of CSCs, which rely on developmental signaling programs.

The heterogeneity of tumor cells has long been appreciated, but two decades ago, seminal work from Dick's laboratory (Bonnet and Dick 1997) described the isolation of a leukemia-initiating cell, the first purification of cancer stem-like cells, a population of cells that had originally been proposed to exist >150 years earlier (Sell 2004). The first prospective isolation of human NSPCs was performed using CD133 (Uchida et al. 2000) and prompted a search for brain tumor cells that shared the characteristics of NSPCs. A burst of studies soon followed describing brain CSCs in anaplastic astrocytoma (Ignatova et al. 2002), medulloblastoma, pilocytic astrocytoma, ependymoma, ganglioglioma (Hemmati et al. 2003; Singh et al. 2003), and GBM (Ignatova et al. 2002; Hemmati et al. 2003; Galli et al. 2004). Brain CSCs have subsequently been shown to be resistant to standard-of-care chemotherapy (Chen et al. 2012) and radiotherapy (Bao et al. 2006a), underscoring their role in disease progression and recurrence.

While cellular heterogeneity within CNS tumors is well recognized (Bonavia et al. 2011; Meacham and Morrison 2013), the nomenclature used to describe the self-renewing population of tumor cells with enhanced tumorigenic properties is far from uniform (see ). To date, many terms have been used to describe this population, including cancer/tumor/glioma/brain tumor stem cell, stem-like tumor cell, cancer-/tumor-/glioma-/brain tumor-initiating cell, and cancer-/tumor-/glioma-/brain tumor-propagating cell. This lack of uniformity has generated confusion and controversy by redirecting the focus away from the biology of these cells and their contribution to tumorigenic processes toward identifying markers that the cells express and whether tumor cells can be propagated as free-floating spheres. In addition, while the term stem cell is used, it does not necessarily mean these cells have been generated from a transformed stem cell, as there is evidence that multiple cell typesranging from stem cell to differentiated progeny, depending on the modelare amenable to oncogenic transformation. Therefore, in the current context, it is essential that the strictest functional assays continue to be performed and a singular term be used for studies using models that fulfill this criteria. As the accepted functional definition of a stem cell is the ability to self-renew and generate differentiated progeny, any claims for a CSC population must also demonstrate this capacity (). For brain tumors, this means the ability to generate a tumor upon intracranial transplantation that recapitulates the cellular heterogeneity present in the parental tumor. Unlike the designation of a tumor-initiating cell, CSCs cannot be investigated in isolation due to the required comparison with differentiated progeny. Prospective enrichment and depletion of tumorigenic and nontumorigenic cells demonstrate the presence of a cellular hierarchy. Cells that meet these criteria (tumorigenic and containing a cellular hierarchy) should be referred to as CSCs (or alternatives such as glioma stem cells, glioma CSCs, or brain tumor stem cells in the context of GBM). While the ability to grow as spheres is also evident in CSCs, it is not by default the defining feature of a self-renewing population of cells. In fact, the majority of spheres derived from both normal and neoplastic brain cells come from progenitor cells with limited self-renewal potential, not stem cells (Pastrana et al. 2011). Furthermore, high-passage cell lines, which do not offer the ability to accurately represent tumor complexity in vivo (Lee et al. 2006), should not be replaced with cells grown in long-term passage as spheres but rather with functionally validated CSC models, as this offers the best opportunity to more deeply model the complexity of brain tumors. Thus, although culture of glioma cells as neurospheres may not be required to maintain stemness (Pollard et al. 2009; Cheng et al. 2012), the microenvironment, including medium composition and culture conditions, does necessarily affect the characteristics of CSCs (Pastrana et al. 2011).

Functional criteria of CSCs. CSCs are defined by functional characteristics that include sustained self-renewal, persistent proliferation, and tumor initiation upon secondary transplantation, which is the definitive functional CSC assay. CSCs also share features with somatic stem cells, including frequency within a tissue (or tumor), stem cell marker expression (examples relevant to GBM and the brain are provided), and the ability to generate progeny of multiple lineages.

Great energy and passion have been devoted to the discovery, validation, and use of CSC enrichment methods. Demonstration of a cellular hierarchy demands methods to separate populations that can be functionally studied. Ideally, an enrichment method would be based on a property that defines an essential CSC feature (self-renewal, tumor initiation, etc.) that is immediately lost upon differentiation (i.e., a digital readout) and is usable with live cells. Currently, no such system exists for any cell type (normal or neoplastic) because biologic systems rarely exhibit all or none phenomena. Critics of the CSC hypothesis have held this limitation up as proof against CSCs; while the same limitations exist for even the best-characterized normal stem cell (hematopoietic stem cell), no scientists deny the existence of hematopoietic stem cells. Leukemia stem cells are considered a definitive tumor population, yet no marker signature for these cells is definitive (Eppert et al. 2011). A more sophisticated and nuanced use of enrichment systems that is informed by recognition of the diversity of GBMs can lead to context-specific methods to produce matched tumorigenic and nontumorigenic populations.

Most glioma CSC markers have been appropriated from normal stem cells, but the linkage between glioma CSCs and normal stem cells remains controversial. Many of the transcription factors or structural proteins essential for normal NSPC function also mark glioma CSCs, including SOX2 (Hemmati et al. 2003), NANOG (Ben-Porath et al. 2008; Suva et al. 2014), OLIG2 (Ligon et al. 2007), MYC (Kim et al. 2010), MUSASHI1 (Hemmati et al. 2003), BMI1 (Hemmati et al. 2003), NESTIN (Tunici et al. 2004), and inhibitor of differentiation protein 1 (ID1) (Anido et al. 2010). However, because of the limited utility of intracellular proteins for enriching CSCs from nonstem tumor cells (NSTCs) using traditional methods such as flow cytometry, a multitude of potential cell surface markers have been suggested, including CD133 (Hemmati et al. 2003), CD15 (also called Lewis x and SSEA-1 [stage-specific embryonic antigen 1]) (Son et al. 2009), integrin 6 (Lathia et al. 2010), CD44 (Liu et al. 2006), L1CAM (Bao et al. 2008), and A2B5 (Ogden et al. 2008). These types of cell surface markers mediate interactions between cells and the microenvironment, but dissociation of cells from their surroundings rapidly degrades the informational content of markers, requiring rapid utilization.

The first proposed marker, CD133 (Prominin-1), a cell surface glycoprotein expressed on neural stem cells, enriches for cells with higher rates of self-renewal and proliferation and increased differentiation ability (Singh et al. 2003). However, CD133 expression, rather than the AC133 surface epitope, should be used with care to enrich for any cells: Surface CD133 marks stem cells and decreases with differentiation, but the expression of Prominin-1 mRNA is not regulated with stemness (Kemper et al. 2010), suggesting that only the glycosylated surface protein CD133 is CSC-specific. The AC133 antigen marks the glycosylated molecule localized in lipid rafts that signals through PI3K and other key pathways to mediate interactions between a cell and its microenvironment (Wei et al. 2013). Most studies fail to recognize this role and use CD133 as a marker in cells that have been extensively cultured out of their microenvironment. Furthermore, the information contained in CD133 is context-dependent. CD133 mRNA, protein lysates, immunofluorescence, and FACS analysis for the AC133 glycoprotein have very different relationships to cell biology. Unfortunately, the complexity of these biomarkers has led to a reductionist view that has challenged the field due to the lack of consistency in methodology and models. It is nearly certain that CD133 is not universally informative in all tumors and has a false-negative rate for identifying CSCs (CD133-negative cells can be tumor-propagating in some tumors) (Beier et al. 2007). Additionally, the use of CD133 as a stem marker is complicated by the observation that expression of CD133 can be regulated at the level of the cell cycle, with potentially slow-cycling NSPCs lacking CD133 expression during G0/G1 cell cycle phase but still maintaining multipotency (Sun et al. 2009).

Although CD133 continues to be the most commonly used cell surface marker, other markers, such as integrin 6, have been proposed to segregate CSCs and NSTCs (Lathia et al. 2010). CD15/SSEA-1 and CD44 have also been proposed as possible markers, potentially with an association with specific subgroups of GBM (Bhat et al. 2013). These markers have utility but must be approached with caution. Each can mark a large percentage of cells, consistent with a high false-positive rate. Due to the current limitations in the functional assays defining CSCs, false-positive markers are sometimes claimed to be superior to functional identification, but markers lack significant utility in discovery studies, which benefit from greater specificity. Additionally, it is likely that no marker will ever be uniformly informative for CSCs because most tissue types contain multiple populations of stem cells expressing different markers and due to the inherent adaptability of cancer cells.

Several methods other than marker expression have been used to enrich for glioma CSCs, such as the abilities to grow as neurospheres in serum-free medium or efflux fluorescent dyes (Goodell et al. 1996; Kondo et al. 2004). Many investigators have used neurosphere culture to select for progenitor cells in the normal and neoplastic brain cells, but there are challenges with this approach. Neurosphere culture selects for a small fraction of the original tumor cells with bias toward progenitor features and expression of epidermal growth factor receptor (EGFR) and FGFR based on growth factors (EGF and FGF) added to the medium (Pastrana et al. 2011). This selection process eliminates the ability to prospectively enrich and deplete stem-like cells, preventing the delineation of a cellular hierarchy required to prove the presence of CSCs. Neurosphere culture selects for cells that can grow in stem cell medium; however, the selection of CSCs simply based on culture methods fails to recapitulate the heterogeneity of the original tumor in vivo as assessed by histological morphology, differentiated cell lineage, and gene expression (Lee et al. 2006; Lathia et al. 2011; Venere et al. 2011), a characteristic that CSCs acutely isolated using marker expression maintain (Singh et al. 2004). An alternative approach to CSC enrichment is the use of flow cytometry to isolate a side population containing CSCs, which is based on the hypothesis that stem cells contain drug efflux transporters (Yu et al. 2008). While this approach has identified a population of self-renewing cells in a mouse glioma model (Bleau et al. 2009), it has not been used successfully to enrich for self-renewing cells in human GBM (Broadley et al. 2011; Golebiewska et al. 2013), highlighting the model- and species-specific challenges of enrichment methods.

CSC markers, although useful to enrich populations of stem cells from nonstem cells, are not sufficient to define either population due to the lack of definitive markers. Functional validationthe observation of differences in stem cell characteristics of CSCs and NSTCsis essential to ensure that the enriched cells truly exhibit the functional characteristics of stem cells (). Various methods, both in vitro and in vivo, are employed to assess stem cell characteristics (self-renewal, proliferation, and ability to reproduce the complexity of the original tumor) of enriched cells. In vitro neurosphere formation assays test for both proliferation and self-renewal but fail to address cellular hierarchy and do not recapitulate the tumor microenvironment. Sphere formation is a surrogate of self-renewal capability andwhen performed in a limiting dilution formatstem cell frequency, but in vivo tumor formation assays are essential to claim the presence of CSCs.

The gold standard for CSC determination remains the ability of a limiting dilution of cells to recapitulate the complexity of the original patient tumor when transplanted orthotopically. The ability to derive heterogeneity is essential because populations of transit-amplifying cells may form a tumor but will only give rise to cells from their specific lineage. Heterotypic transplantation of cellsfor example, into the flank of the animalmay also be informative, but this technique lacks the proper microenvironmental cues of orthotopic implantation.

Glioma CSCs are regulated by six main mechanisms, which include intrinsic factors such as genetics, epigenetics, and metabolism as well as extrinsic qualities of niche factors, cellular microenvironment, and the host immune system (). The following sections describe the key features of each of these factors and highlight new advances in the topics of epigenetics mapping, single-cell heterogeneity, metabolism, and immunotherapy.

Regulation of CSCs. Cell-autonomous (intrinsic) and external (extrinsic) forces regulate the CSC state. Key intrinsic regulators include genetic, epigenetic, and metabolic regulation, while extrinsic regulators include interaction with the microenvironment, including niche factors and the immune system.

Through advances in genomic technologies, we now have a comprehensive picture of the genetic mutations and structural variants present in GBM (Atlas 2008; Brennan et al. 2013). Some of the most recurrent alterations include EGFR, IDH1, PDGFRA, HDM2, PIK3CA, and TERT promoter and PI3KR1 gain-of-function mutations or amplifications and mutations or deletions of the tumor suppressors PTEN, TP53, CDKN2A, NF1, ATRX, and RB1. While many of these mutations are prevalent in several other cancer genomes, several mutations are highly enriched in GBM, such as IDH1 mutations, which lead to a CIMP (G-CIMP) (Noushmehr et al. 2010). These studies highlight the significant degree of intertumoral heterogeneity present in GBM, which is further captured at both the transcriptional and epigenetic levels (Phillips et al. 2006; Verhaak et al. 2010), and also underscore the complexity of the clonal evolution and clonal diversity that occur during the genesis of GBM and their bearing on the shape and structure of the CSC hierarchy. While both genetic and epigenetic landscapes define functionally distinct clones during tumor evolution, epigenetic differences likely account for the functional differences between cells within the hierarchy.

Epigenetic maintenance of the CSC state is regulated largely at the level of transcriptional and chromatin regulation. CSC regulation converges on MYC, which occurs in the presence of MYC-mediated cancer cell survival and proliferation programs (Wang et al. 2008; Zheng et al. 2008; Wurdak et al. 2010; Chan et al. 2012; Fang et al. 2014). Additional transcription factors have been identified as important for CSC identity, including STAT3 (Sherry et al. 2009), SOX2 (Gangemi et al. 2009), FOXM1 (Joshi et al. 2013), FOXG1 (Verginelli et al. 2013), GLI1 (Clement et al. 2007), ASCL1 (Rheinbay et al. 2013), ZFX (Fang et al. 2014), NANOG (Zbinden et al. 2010), and ZFHX4 (Chudnovsky et al. 2014), which recruit necessary chromatin remodeling factors to promote maintenance of the glioma CSC state. By using epigenome-wide mapping of cellular chromatin state, Suva et al. (2014) identified a core set of four transcription factors in proneural GBM able to reprogram differentiated tumor cells into glioma CSCs. These investigators showed that POU3F2, SOX2, SALL2, and OLIG2 are master transcription factors required to maintain the tumor-forming capability of these cells, suggesting that mediators of stem cell programs could capture the oncogenic capacity of CSCs. In addition to transcription factors, regulators of nucleosome structure have also been reported to maintain the CSC state. The mixed lineage leukemia 1 (MLL1) protein has been shown to maintain the CSC phenotype through activation of HOXA10, which subsequently regulates a network of homeobox genes that is required for tumor maintenance (Heddleston et al. 2012; Gallo et al. 2013). Similarly, the H3K27 methylase EZH2 has been shown to be important for CSC maintenance through its function as a regulator of both Polycomb-repressive domains and STAT3 signaling (Kim et al. 2013). The BMI1 Polycomb ring finger oncogene regulates both normal neural stem cells and GBM cells (Bruggeman et al. 2007).

These studies highlight the importance of understanding the dynamics of core transcription factors in maintaining stem cell state and the effect that these factors have on shaping the chromatin landscape of cells within the tumor hierarchy.

Single-cell RNA sequencing (RNA-seq) interrogation of cellular heterogeneity within GBMs identified novel genes predominantly present in GBM CSCs compared with differentiated cells and provocatively detected cells of multiple GBM subtypes within single tumors, drawing into question the utility of subtyping tumors and targeting specific subtypes (Patel et al. 2014). Furthermore, these investigators described an inverse correlation between stem signature and cell cycle gene expression, suggesting that the cells that form neurospheres in culture cycle more slowly compared with differentiated and differentiating tumor cells. A parallel, single-cell functional analysis of GBMs confirmed a strong variation of genomics and response to therapy (Meyer et al. 2015). Additional detailed analysis of heterogeneity of this type will greatly expand our understanding of the differences between tumor cells both within and among GBM patients and improve the characterization of glioma CSCs.

GBM CSCs reside in varied tumor microenvironments that limit nutrients, such as glucose and oxygen. Under such conditions, cancer cells, including glioma CSCs, exhibit the Warburg effect, a metabolic shift toward aerobic glycolysis and the accumulation of lactate in exchange for sustained ATP production and metabolite generation for macromolecule synthesis. Glioma CSCs demonstrate plasticity in the metabolic pathways used in response to metabolic restrictions and may shift toward the use of the pentose phosphate shunt (Vlashi et al. 2011; Kathagen et al. 2013). This inherent persistence of CSCs under hypoxic and acidic conditions as well as the preferential utilization of HIF-2 signaling compared with NSTCs and normal progenitors promote the maintenance of self-renewal, proliferation, and survival (Li et al. 2009b). Similarly, in conditions of nutrient deprivation such as low glucose, glioma CSCs outcompete neighboring NSTCs for glucose uptake through preferential up-regulation of the high-affinity GLUT3 transporter (Flavahan et al. 2013). A consequence of altered metabolic state is the production of reactive oxygen species. Glioma CSCs not only are dependent on NOS2 activity for promoting tumor growth but also synthesize nitric oxide through the specific up-regulation of NOS2 protein (Eyler et al. 2011). Importantly, in GBM, cellular metabolic characteristics are often genetically hardwired, such as recurrent IDH1 mutations, which are commonly observed in proneural GBM. Mutant IDH1 leads to a gain-of-function enzymatic activity, causing accumulation of 2-hydroxyglutarate, an oncometabolite that inhibits the TET1 and TET2 demethylases to cause aberrant hypermethylation of DNA and histones. While the function of IDH1 mutations in the context of CSCs is not directly defined, IDH1 mutations induce a loss of differentiation, preventing the terminal differentiation of lineage-specific progenitors (Lu et al. 2012). Moving forward, integrated metabolomic and epigenomic profiling may reveal other examples of intricate relationships between metabolism and epigenetic programs and their influence on the glioma CSC state.

Brain development is orchestrated by a series of regulatory pathways with spatially and temporally controlled activity. Notch and NF-B (nuclear factor B) signaling instructs the fate of NSPCs, with the guidance and lineage commitment of progeny dictated by pathways that include the ephrins and bone morphogenetic proteins (BMPs). In a manner that mimics aberrant differentiation, CSCs co-opt developmental programs to maintain an undifferentiated state, increasing their survival and maintenance. Common pathways activated in CSCs include Notch, BMP, NF-B, and Wnt signaling (Li et al. 2009a; Day et al. 2013; Rheinbay et al. 2013; Lubanska et al. 2014; Yan et al. 2014). Collectively, niche factors represent an overriding theme in CSC biology, where stem and progenitor cell features provide selective advantages to maintain tumor growth (). These pathways may be activated through a combination of genetic and epigenetic alterations in addition to microenvironmental and metabolic factors.

The Notch pathway plays a role during neural development, functioning to inhibit neuronal differentiation and sustain NSPC populations. This pathway is co-opted in GBM, where aberrant NOTCH activation stimulates astrocytes to assume a stem-like state accompanied by increased proliferation (Jeon et al. 2008). The importance of Notch signaling in glioma CSC biology is highlighted by the convergence on this pathway from other pathways and exogenous factors, such as hypoxia, eNOS signaling, and response to radiation (Charles et al. 2010; Wang et al. 2010; Qiang et al. 2012). The dependence of glioma CSCs on Notch signaling is further supported by experiments demonstrating depletion of CSCs by treatment with -secretase inhibitors (Fan et al. 2006, 2010).

As BMPs direct NSPC fate toward an astroglial lineage, these signals have been proposed as a possible differentiation therapy for GBM (Piccirillo et al. 2006). Despite the presence of BMP expression in primary GBM tissue, glioma CSCs are highly resistant to the differentiation effects of BMPs in a process that occurs through at least two distinct cell-autonomous mechanisms: the shift to a fetal BMP receptor expression in glioma CSCs through recruitment of the transcriptional repressor EZH2 (Lee et al. 2008) and the secretion of BMP antagonists, specifically Gremlin1, by CSCs to protect against endogenous BMP-mediated differentiation (Yan et al. 2014). In this manner, CSCs generate differentiated progeny that provide supportive cues to the parental cells (e.g., Notch ligands, interleukin-6 [IL-6], and extracellular matrix) while resisting differentiation signals.

The NF-B pathway has emerged as an important regulator of GBM cell survival and identity through an endogenous cell stress response transcriptional program (Bhat et al. 2013). The A20 protein (TNFAIP3), a mediator of cell survival and the NF-B pathway, is overexpressed in CSCs compared with NSTCs (Hjelmeland et al. 2010). Supporting these findings, Sema3C and its receptors, PlexinA2 and PlexinD1, are also coordinately expressed in CSCs and activate Rac1 and NF-B in an autocrine/paracrine loop to promote CSC survival (Man et al. 2014).

GBM CSCs have also been shown to be highly dependent on Ephrin receptor signaling for survival and the maintenance of stem cell properties. Specifically, Ephrin A molecules and the EPHA2 and EPHA3 receptors are highly expressed in glioma CSCs and potentially function through the negative regulation of mitogen-activated protein kinase (MAPK) signaling (Binda et al. 2012; Day et al. 2013).

Wnt signaling is highly active in CSCs and is critical for the maintenance of the stem cell phenotype. An integrated genomic and biological analysis identified PLAGL2 as highly amplified in gliomas with functional suppression of CSC differentiation through modulation of Wnt/-catenin signaling (Zheng et al. 2010). Comprehensive mapping of chromatin modifications in CSCs and their NSTC counterparts revealed widespread activation of Wnt pathway genes through loss of Polycomb-mediated repression. The CSC chromatin landscape is thought to be dependent on achaete scute family basic helixloophelix (bHLH) transcription factor 1 (ASCL1), which activates Wnt signaling through negative regulation of dickkopf WNT signaling pathway inhibitor 1 (DKK1) (Rheinbay et al. 2013). Hedgehog signaling in the CNS is mediated in part by NSPC communication with the cerebrovascular fluid through primary cilia. Gliomas contain primary cilia, and the resulting CSCs are dependent on hedgehog signaling (Bar et al. 2007; Clement et al. 2007; Ehtesham et al. 2007).

Given the role of growth factors in normal brain development, it is not unexpected that numerous canonical growth factor signaling pathways have been shown to contribute to GBM maintenance and function. PDGFR signaling promotes CSC survival, self-renewal, and invasion and tumor growth through downstream STAT3 activation (Kim et al. 2012). Similarly, glioma CSCs preferentially express the IL-6 receptor, which also promotes convergent signaling upon STAT3 activation (Wang et al. 2009).

EGFR signaling has also been reported to contribute to CSC maintenance through the activation of AKT, the recruitment of SMAD5, and the induction of ID3, IL-6, and IL-8. This suggests a potential hypothesis in which the EGFR and PDGFR pathways are linked by IL-6 signaling. A potential alternate hypothesis is the presence of distinct CSC populations dependent on different growth factor receptor signaling pathways. Supporting this latter notion, EGFR inhibition promotes expansion of a cMET growth factor receptor-positive population of CSCs (Jun et al. 2014). Furthermore, elevated cMET expression is important for CSC maintenance, tumorigenicity, and resistance to radiation (Joo et al. 2012).

Aligned with its role in stress responses, transforming growth factor (TGF-) stimulates CSC self-renewal. Autocrine TGF- signaling permits retention of stemness through positive regulation of SOX2 and SOX4 expression (Ikushima et al. 2009). A distinct subset of TGF--dependent CSCs expresses CD44 and ID1 (Anido et al. 2010), which are markers of functionally distinct CSCs. A crucial mediator of the TGF- response in CSCs is the BMI1 protein, which connects stem cell programs and ER stress pathways through the transcriptional repressor ATF3 (Gargiulo et al. 2013).

Immune suppression is a hallmark of cancer (Hanahan and Weinberg 2011); while the brain possesses a unique series of immune surveillance mechanisms that become active during pathogenic states (Ransohoff and Engelhardt 2012), brain tumors have been characterized as immunosuppressive (Platten et al. 2001; Fecci et al. 2006). There is increasing enthusiasm for immunotherapy strategies based on the limited success of signaling pathway inhibitors and anti-angiogenic agents in brain tumors and the success of immunotherapy in melanoma. Immunotherapies for brain tumors include cellular (adoptive T-cell transfer and chimeric antigen receptor engineered T cells), vaccination, and immunomodulatory therapies targeting immune checkpoints (including anti-programmed death 1 [PD1], PD ligand 1 [PD-L1], and cytotoxic T lymphocyte-associated protein 4 [CTLA4] antibodies) (Reardon et al. 2014). Reversing tumor-induced immune suppression by increasing cytotoxic cell function and reducing suppressor cell function may unleash the endogenous immune response. Immunologic therapies may offer an additional benefit, as most strategies do not require intracranial delivery, a major restriction point for many oncologic treatments. While CSCs are key drivers of tumor growth, CSC interactions with the immune system and potential exploitation in immunotherapy are under active investigation (). These studies will require innovative approaches, as the majority of CSC studies involve xenograft models that lack major immune cell components, and many mouse models have reduced cellular heterogeneity. However, the information obtained from mouse model approaches is likely to be informative for the human immune response, as genetically engineered mouse models can recapitulate key aspects of brain tumor immunosuppression (Kong et al. 2010).

Proposed features of CSCs. Non-cell-autonomous aspects of CSCs may drive tumor growth but also serve as points of fragility. These include the increased ability to invade through the brain parenchyma, immune evasion, relationship with a niche, and promotion of angiogenesis.

Despite these challenges, there is building evidence that CSCs directly modulate the immune system. In coculture studies, CSCs induced regulatory T cells while inhibiting proliferation and cytotoxic T-cell activation with a concomitant induction of cytotoxic T-cell apoptosis, mediated via PD1 and soluble galectin-3 (Di Tomaso et al. 2010; Wei et al. 2010). Other CSC-secreted factors include IL-10 and TGF-, which also suppresses tumor-associated microglia/macrophage function and generates a more immunosuppressive (M2) phenotype (Wu et al. 2010). Another immunotherapy approach that may benefit from CSC targeting is the development of anti-tumor vaccines. Current vaccine efforts have focused on tumor-specific antigens (such as EGFRvIII) or whole tumor cell lysates, and there is evidence from preclinical models that CSC lysates are more effective in generating dendritic cell (DC) vaccines than differentiated cells (Pellegatta et al. 2006; Xu et al. 2009). CSCs modulate T-cell and tumor-associated microglia/macrophage function through secreted factors (Zhou et al. 2015), which may be exploited in the development of vaccine strategies or in combination with other drugs (Sarkar et al. 2014). These data provide a rationale for future studies investigating how the interaction between CSCs and other immune cell populations may drive immune suppression and in vivo interrogations into how CSC targeting may alter the immune activation status. Evaluating changes in CSC populations as a result of immunotherapy will also be essential, as will be evaluating combinatorial targeting strategies using immunotherapies and anti-CSC approaches.

Most conventional anti-neoplastic therapies target proliferating cells, but the malignancy of advanced cancers also derives from effects on the immune system, vasculature, and invasion/metastasis (). GBMs infiltrate the surrounding brain, precluding curative surgical resection. Infiltrative tumors must adapt to new environments, including the formation of new vessels to obtain nutrients. GBMs express proangiogenic growth factors (Batchelor et al. 2007), with CSCs driving neoangiogenesis with high levels of VEGF (Bao et al. 2006b). The humanized monoclonal antibody bevacizumab was developed to target VEGF to inhibit angiogenesis and has been used to treat recurrent GBM (Cohen et al. 2009). Bevacizumab attenuates tumor size, but the surviving tumor may display increased invasion in human and mouse models (de Groot et al. 2010), potentially due to a release of c-MET inhibition (Lu et al. 2012). Cancer cells often activate redundant angiogenic pathways in response to VEGF pathway inhibition (Atlas 2008). CSCs located at the perivascular niche are in close contact with the endothelial cells (Calabrese et al. 2007), permitting engagement of endothelial cell Notch ligands with glioma CSC Notch receptors to activate Notch signaling, which supports self-renewal of glioma CSCs (Zhu et al. 2011). CSCs also contribute to vascular structure through transdifferentiation into pericytes to promote tumor growth (Cheng et al. 2013). Inhibition of CSC-derived pericytes disrupts angiogenesis and inhibits tumor growth, directing attention toward nonendothelial cell targeting strategies. Anti-angiogenic drugs in current use have failed to provide a significant survival benefit to GBM patients (Gilbert et al. 2014), suggesting that a benefit may exist to investigating the mechanisms by which tumor cells regulate angiogenesis and that contribute to tumor growth and maintenance to efficiently target the GBM vasculature.

The mainstay treatment of GBM involves surgery, concurrent radiation with chemotherapy, and adjuvant chemotherapy with TMZ (Stupp et al. 2009). Despite advances in the field, the overall survival rate remains only 1519 mo (Stupp et al. 2009). The high degree of tumor heterogeneity in GBM contributes to treatment failure, to which functional and molecular heterogeneity and aberrant receptor tyrosine kinase (RTK) activity all contribute. CSCs located at the top of the hierarchy initiate and maintain the tumor after treatment (Chen et al. 2012). Glioma CSCs have also been shown to contribute to radiation resistance by increasing the DNA damage response machinery (Bao et al. 2006a). In terms of molecular heterogeneity, different subtypes of GBM with distinct molecular profiles coexist within the same tumor and likely exhibit differential therapeutic responses (Sottoriva et al. 2013). For example, several RTKs, including PDGFR in the proneural and EGFR in the classical subtype, are altered in GBM (Verhaak et al. 2010). The abnormal activation of RTKs involves many pathways that are redundant and can initiate and maintain downstream signaling, making tumors refractory to treatment (Stommel et al. 2007). A recent single-cell analysis of primary GBM patients showed that cells from the same tumor have differential expression of genes involved in oncogenic signaling, proliferation, immune response, and hypoxia (Patel et al. 2014). Furthermore, an increase in tumor heterogeneity was associated with a decrease in patient survival. The addition of TMZ to radiation has increased median survival by several months (Stupp et al. 2009), but lineage tracing studies in mouse models demonstrate that CSCs repopulate brain tumors after TMZ treatment (Chen et al. 2012). A number of molecular mechanisms have been identified that mediate the therapeutic resistance of CSCs to cytotoxic therapies, including the DNA damage checkpoint, Notch, NF-B, EZH2, and PARP (Bao et al. 2006a; Wang et al. 2010; Bhat et al. 2013; Venere et al. 2014; Kim et al. 2015), which suggests that CSCs develop multiple mechanisms of resistance that may require combinations of targeted agents. Moving forward, these studies demonstrate the importance of understanding the molecular alterations that are present in recurrent tumors and how these influence the structure of cells within the tumor hierarchy. In addition, it is necessary to consider that therapeutic resistance mechanisms may not be solely innate but may evolve from exposure to microenvironmental factors such as hypoxia and acidic and metabolic stress (Heddleston et al. 2009; Li et al. 2009b; Hjelmeland et al. 2011; Flavahan et al. 2013; Xie et al. 2015).

Conventional treatment for GBM promotes a transient elimination of the tumor and is almost always followed by tumor recurrence, possibly with an increase in the percentage of CSCs (Auffinger et al. 2014), as CSCs are involved in tumor recurrence and therapeutic resistance (Bao et al. 2006a; Chen et al. 2012). To effectively eliminate CSCs, it is critical to target their essential functions and their interactions with the microenvironment. Treatment with TMZ may kill CSCs that contain higher expression of the DNA repair protein MGMT; however, TMZ cannot prevent self-renewal of CSCs that contain MGMT (Beier et al. 2008). Another feature of CSCs is their ability to evade apoptosis. A potential therapeutic strategy would be the use of PARP inhibitors to enhance apoptosis under genotoxic damage. When the PARP inhibitor ABT-888 was used in combination with TMZ and radiation in GBM cell lines, apoptosis increased, and cells were sensitized to therapy (Barazzuol et al. 2013). GBMs thrive in harsh microenvironments characterized by hypoxia and limited nutrient availability. The HIF family of transcription factors is involved in promoting angiogenesis and cell migration in hypoxic regions (Kaur et al. 2005), and several drugs have been developed to target this gene family, with a few undergoing clinical trials. For example, as described previously, glioma CSCs reprogram their metabolic machinery and preferentially take up glucose to survive in environments with limited nutrients by expressing the high-affinity glucose transporter GLUT3 (Flavahan et al. 2013). GLUT3 therefore represents a promising therapeutic target for potential selective inhibition of CSCs. Epigenetic modifications are manifest in tumor recurrence (Nagarajan and Costello 2009). Histone acetylation and methylation are reversible and can be targeted by drugs; the histone deacetylase (HDAC) inhibitor vorinostat is currently in clinical trials (Bezecny 2014). Immunotherapy is an additional emerging therapeutic approach for GBM. The development of vaccines based on heat-shock proteins, EGFRvIII (Del Vecchio et al. 2012), and DCs (Terasaki et al. 2011) has shown promising results in clinical trials. ICT-107, a patient-derived DC vaccine developed against six antigens highly expressed in glioma CSCs (Phuphanich et al. 2013), is currently under clinical evaluation for use in patients.

Some of the challenges of developing therapeutic targeting agents are derived from the lack of universally informative markers to identify CSCs and the common molecular pathways shared by CSCs and NSPCs. The understanding of the biology of the CSCs and how these cells interact with their microenvironment in combination with the genetic and epigenetic landscape in GBM will be essential to develop more effective therapies.

As biological observations have revealed increasing levels of complexity, mathematical modeling approaches have provided a framework to understand the dynamic complexity of stem cell self-renewal and differentiation. By use of proliferation data and lineage tracing analysis, quantitative models have been generated for tissue-specific stem cells that have provided insight into the kinetics of cell fate choice and tissue development (Blanpain and Simons 2013). Similar approaches have been taken to reduce the complexity of CSCs. A network-based model has suggested that CSCs can transition between plastic (proliferative, symmetrically dividing, and less invasive) and rigid (quiescent, asymmetrically dividing, and more invasive) networks that can be modulated by extrinsic stressors, such as hypoxia, inflammation, and therapies (Csermely et al. 2015). Testing this model with biological data is likely to provide additional insights into the complexity of CSCs and identify points of fragility for additional therapeutic development. Mathematical approaches have also been used to evaluate the dynamics of GBM growth. Proliferation and invasion are phenotypes that have been modeled (Harpold et al. 2007). By use of a model that takes into account rates of proliferation and invasion in combination with imaging data, it has been proposed that IDH1 mutant tumors are actually less proliferative and more invasive (Baldock et al. 2014). Clinically relevant parameters, such as identifying optimal radiation schedules, have also been modeled using genetically engineered mice (Leder et al. 2014). Additionally, quantitative approaches have been developed to model the events leading to intertumoral and intratumoral heterogeneity in both human patient specimens (Sottoriva et al. 2013) and mouse models (Cheng et al. 2012). Integrating mathematical approaches into future CSC studies will provide an opportunity to identify key pathways essential for self-renewal and will predict responses to therapeutic perturbations.

GBM provides an excellent system to investigate the importance of CSCs. While there is a standard set of assays used to enrich for and identify CSCs, it remains unclear whether multiple CSC populations exist in different niches (perivascular and hypoxic) and possess different characteristics (slow vs. rapid cycling) as well as how key developmental signaling pathways are used by each of these populations. In addition, while a hierarchy is in place for GBM, the current view of CSCs and NSTCs is mutually exclusive and lacks a progenitor cell population that serves as an intermediate for differentiated progeny generation from somatic stem cells. Mouse studies have revealed that multiple stem and progenitor cell populations have the capacity to give rise to tumors upon oncogenic transformation, but it remains unclear whether there is a single cell of origin for the human disease or, more likely, whether multiple cells of origin exist and how this may be linked to genetic diversity. Making inroads into these unresolved questions will refine the experimental foundation upon which translational studies aiming to develop novel anti-CSC therapies are built and provide key signaling pathways responsible for CSC maintenance that are amenable for targeting.

The extensive molecular characterization of gliomas of all grades has permitted the recognition that the continuum of tumor grade has hidden a set of genetically distinct diseases. IDH1 mutations produce an oncometabolite, 2-hydroxyglutarate, that reprograms cellular chromatin to assume a stem-like state (Lu et al. 2012). Thus, IDH1 mutant gliomas may have a relatively flat hierarchy, with most tumor cells acquiring stem-like features early in tumor initiation. In contrast, primary GBMs accumulate a greater diversity of genetic and epigenetic alterations, which is associated with a more vertical cellular hierarchy. This duality of tumor biology resembles that of the two forms of head and neck cancers. Human papilloma virus-induced head and neck cancers are morphologically uniform and, like IDH1 mutant gliomas, are more responsive to therapies. Alcohol- and tobacco-associated head and neck cancers harbor more mutations and display a worse outcome with a reliable cellular hierarchy. Large-scale genomic sequencing has informed commonalities among cancer types based on driving genetic lesions. It is possible that similar patterns will be appreciated with cancer types based on epigenetic and cellular hierarchies, creating broader opportunities to improve diagnostics and therapeutics. In fact, expanding the organizational structures is likely to be a useful approach to increase our understanding of complex disease states. Many diseases display heterogeneous aspects that are governed by both cell-autonomous and microenvironmental forces. With the success of immunotherapy approaches to activate the immune system via immune checkpoint inhibition in cancers such as melanoma, understanding how GBM and, in particular, CSCs interface with the immune system is an immediate priority. An alternative view of heterogeneity and therapeutic response may also be informative for future studies. For example, bacterial infections contain distinct populations of cells that have different proliferative potential and responses to therapy. Viable but nonculturable bacteria and latent infections, including tuberculosis, may be found in particular niches associated with inflammation, hypoxia, acidic and nitrosative stress, and nutrient restriction (Oliver 2010). Most antibiotics, like anti-neoplastic agents, are directed against the proliferative population, leaving a resistant population behind. Novel methods are being used to screen for new agents that target resistant bacteria, such as latent tuberculosis (Bryk et al. 2008). Nathan (2004) suggested that essentiality is conditional, and the conditions defining essentiality are multiple in the context of latent infections. An identical view can instruct CSC targeting efforts as we grow in our understanding of the governing stimuli both internal and external to CSCs.

One infrequently discussed point is a re-equilibration of a cellular hierarchy in tumors generated from CSCs. If cell-autonomous advantages were the sole determinant of the differentiation state of tumor cells, CSCs would represent the majority of tumor cells, as the evolutionary drive toward increased fitness would provide a selective advantage to CSCs. At steady state (in distinction from homeostasis), tissues balance competing requirements through multiple levels of interaction among stem cells, progenitor cells, and differentiated progeny. Collectively, the individual cellular dynamics in cancer permits tumors to respond to exogenous insults (cytotoxic therapies, immunologic attack, etc.) to maintain the aberrant organ system. These dynamics are also at play within the cellular hierarchy in which CSCs give rise to NSTCs, and, when necessary, NSTCs give rise to CSCs to maintain the cellular equilibrium necessary for optimal tumor growth. CSCs should not be considered a model to simplify the modeling of GBMs and other cancers, but rather the CSC hypothesis constitutes an additional level of complexity that contributes to the malignancy of cancers. As CSCs reside in multiple niches governed by different molecular programs, there will not be single anti-CSC targeted therapeutics with broad activity; instead, CSCs will demand multitargeted approaches. Patients with GBMs are in desperate need of improved therapies. The real validation of CSCs will come with better treatments due to the integration of CSCs into drug development.

We sincerely apologize to those investigators whose work we were unable to cite due to space limitations. We thank Amanda Mendelsohn (Center of Medical Art and Photography, Cleveland Clinic) for assistance with figure preparation. We also thank our funding sources: The National Institutes of Health (grants CA154130, CA171652, CA169117, NS087913, and NS089272 to J.N.R., and CA157948, CA191263, and NS083629 to J.D.L.); Sontag Foundation (J.D.L.); Research Programs Committees of Cleveland Clinic (J.N.R); and James S. McDonnell Foundation (J.N.R). S.C.M. is supported by a Canadian Institutes of Health Research Banting Fellowship. Work in the Lathia laboratory is also supported by the Lerner Research Institute, Case Comprehensive Cancer Center, Voices Against Brain Cancer, Blast GBM, the Ohio Cancer Research Associates, Research Scholar Award from the American Cancer Society, V Scholar Award from the V Foundation for Cancer Research, and grant IRG-91-022-18 to the Case Comprehensive Cancer Center from the American Cancer Society.

Original post:
Cancer stem cells in glioblastoma

Posted in Ohio Stem Cells | Comments Off on Cancer stem cells in glioblastoma

Page 533«..1020..532533534535..540550..»