header image

Page 28«..1020..27282930..4050..»

Archive for Pet Stem Cell Therapy

Ask the Doc: Could I get blood cancer from exposure to harmful pesticides? – The Straits Times

Which is the most prevalent blood cancer in Singapore, and what are common signs and symptoms to look out for?

The most prevalent blood cancer is lymphoma. Blood cancers usually originate in the bone marrow, lymph nodes, or various organs. They can also spread to other organs. Symptoms are related to the specific organ affected; for example, there could be abdominal swelling and pain if the liver and spleen are enlarged.

Common symptoms include fever, weight loss, night sweats, bleeding, fatigue, a sickly pallor, lymph node swelling, bone and joint pain.

Blood cancers can be divided into three categories:

Acute or AggressiveFrom the onset, acute leukaemia and aggressive or high-grade lymphoma progress rapidly in a matter of a few weeks to a few months. Without treatment, death is almost always imminent.

ChronicChronic leukaemia, indolent/ low-grade lymphoma and multiple myeloma can go undetected over a long period of time from many months to years until the patient becomes symptomatic. Some conditions can be monitored without being treated, till symptoms and indications surface.

Pre-cancerousMyelodysplastic syndrome and myeloproliferative diseases are blood disorders that can vary from mild to severe conditions. Mild disorders can remain dormant for years, yet progress to severe conditions or transform into acute leukaemia.

While the exact cause of blood cancers is generally unknown, associated risk factors have been identified for specific diseases.

Acute myeloid leukaemia is known to be associated with previous exposure to agents toxic to cells (like chemotherapy and radiotherapy for previous cancers); harmful chemicals and pesticides; and underlying chronic blood disorders such as myelodysplastic syndrome and myeloproliferative neoplasms.

Some blood cancers are also linked to inherited and acquired genetic mutations, or chronic viral conditions such as the Human Immunodeficiency Virus or Epstein Barr Virus.

Ethnic variation can also be found in the prevalence of some blood cancers. For example, chronic lymphocytic leukaemia is more prevalent in Caucasians but T-cell lymphoma is more prevalent in Asians.

Apart from blood tests,blood grouping may also be necessary at the diagnosis stage becausepatients frequently need a blood transfusion. PHOTO: GETTY IMAGES

Mainly by detecting cancer cells in involved tissues or organs.

Bone marrow tests can be carried out to detect cancer cells originating from there. The tests will also detect the spread of cancer cells from other parts of the body to the bones. For instance, lymphoma usually originates in the lymphoid tissue and can spread to other parts of the body, including bones.

Blood tests can detect cancer cells present in a patient's blood. Basic screening tests including kidney and liver function are done, prior to treatment. Infectious disease screening can be done to uncover associated infections, and blood grouping may also be necessary at the diagnosis stage becausepatients frequently need a blood transfusion.

Imaging studies, computerised tomography (CT) scan, positron emission tomography (PET-CT scan) and magnetic resonance imaging (MRI) scans can also evaluate the stage of blood cancers (usually in lymphoma).

A lumbar puncture and or cerebrospinal fluid examination may also be performed to evaluate if blood cancer has spread to the brain and the spinal cord.

Aimed at achieving durable remission, leading to the cure of the disease, treatment options vary depending on the type of blood cancers.

A combination of chemotherapy, chemoimmunotherapy and/or targeted drugs are used to induce remission in blood cancers. Subsequently, further rounds of the same treatments, or consolidation with peripheral blood or bone marrow stem cell transplantation will be done. Sometimes, radiotherapy is used as a consolidation therapy. Some patients may also need maintenance therapy.

Should there be a relapse, salvage therapy can be done with available conventional treatment options, or via clinical trials. Newer treatment options for relapsed diseases include immunotherapy like Bispecific T-cell Engagers and cell therapy like Chimeric Antigen Receptor T-cells.

Cancer cells survive by suppressing or bypassing the immune system. T-cells are a type of immune cells that keep abnormal cells in check, and kill cancer cells. While a patients T-cells cannot recognise or kill cancer cells on their own, they can be modified into chimeric antigen receptor (CAR) T-cells.

It involves altering the genes inside T cells to help them attack cancer cells, and has been approved by the U.S. Food and Drug Administration for use among a specific group of patients.

Anti-CD19 CAR-T treatment can be delivered for relapsed acute lymphoblastic leukaemia in children and adults aged below 25 (recently approved in Singapore), Anti-CD19 CAR-T treatment is also available for relapsed diffuse large B-cell lymphoma, and anti-BCMA CAR-T treatment addresses relapsed multiple myeloma.

Potential side effects such as neurological complications and cytokine release syndrome as a result of overwhelming immune activation can be expected, so a multidisciplinary team approach among experts is necessary to help manage patients undergoing this type of treatment.

Read the original here:
Ask the Doc: Could I get blood cancer from exposure to harmful pesticides? - The Straits Times

Incyte and MorphoSys Announce First Patient Dosed in Phase 3 inMIND Study Evaluating the Addition of Tafasitamab to Lenalidomide and Rituximab in…

WILMINGTON, Del. & PLANEGG, Germany & MUNICH--(BUSINESS WIRE)--Incyte (Nasdaq:INCY) and MorphoSys AG (FSE: MOR; Prime Standard Segment; MDAX & TecDAX; NASDAQ:MOR) today announced the first patient has been dosed in the placebo-controlled Phase 3 inMIND study evaluating the efficacy and safety of tafasitamab or placebo in combination with lenalidomide and rituximab in patients with relapsed or refractory follicular lymphoma (FL) or marginal zone lymphoma (MZL).

Despite improvements in treatment for patients with relapsed or refractory FL and MZL, there continues to be a significant medical need for additional therapies with improved outcomes, said Peter Langmuir, M.D., Group Vice President, Oncology Targeted Therapeutics, Incyte. We are pleased to have initiated the inMIND study as we seek meaningful, new options for patients with relapsed or refractory FL or MZL.

FL and MZL are the most common indolent, or slow growing, forms of B-Cell non-Hodgkin lymphomas (NHLs). FL and MZL account for approximately 20-25% and 7% of adult NHL cases, respectively.1 There are limited treatment options for the more than 17,000 new cases of relapsed or refractory FL treated every year in the United States, Europe and Japan.2

"We are looking forward to building on previous, exploratory data in FL, and the results seen with tafasitamab and lenalidomide in relapsed or refractory diffuse large B-cell lymphoma, to evaluate the potential benefit of adding tafasitamab to the current lenalidomide and rituximab combination regimen in patients with indolent lymphomas, said Mike Akimov, M.D., Ph.D., Head of Global Drug Development, MorphoSys.

On January 7, 2021, the U.S. Food and Drug Administration granted orphan drug designation to tafasitamab for the treatment of FL.

About inMIND

inMIND (NCT04680052), a global, double-blind, placebo-controlled, randomized Phase 3 study, is evaluating whether tafasitamab and lenalidomide as an add-on to rituximab provides improved clinical benefit compared with lenalidomide alone as an add-on to rituximab in patients with relapsed or refractory follicular lymphoma (FL) Grade 1 to 3a or relapsed or refractory nodal, splenic or extranodal marginal zone lymphoma (MZL). The study is expected to enroll over 600 adult (age 18 years) patients with relapsed or refractory FL or MZL.

The primary endpoint of the study is progression-free survival (PFS) in the FL population, and the key secondary endpoints are PFS and overall survival (OS) in the overall population as well as positron emission tomography complete response (PET-CR) at the end of treatment (EOT) in the FL population.

For more information about the study, please visit: https://clinicaltrials.gov/ct2/show/NCT04680052

About Tafasitamab

Tafasitamab is a humanized Fc-modified cytolytic CD19 targeting monoclonal antibody. In 2010, MorphoSys licensed exclusive worldwide rights to develop and commercialize tafasitamab from Xencor, Inc. Tafasitamab incorporates an XmAb engineered Fc domain, which mediates B-cell lysis through apoptosis and immune effector mechanism including antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP).

Monjuvi (tafasitamab-cxix) is approved by the U.S. Food and Drug Administration in combination with lenalidomide for the treatment of adult patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) not otherwise specified, including DLBCL arising from low grade lymphoma, and who are not eligible for autologous stem cell transplant (ASCT). This indication is approved under accelerated approval based on overall response rate. Continued approval for this indication may be contingent upon verification and description of clinical benefit in a confirmatory trial(s).

In January 2020, MorphoSys and Incyte entered into a collaboration and licensing agreement to further develop and commercialize tafasitamab globally. Monjuvi is being co-commercialized by Incyte and MorphoSys in the United States. Incyte has exclusive commercialization rights outside the United States.

A marketing authorization application (MAA) seeking the approval of tafasitamab in combination with lenalidomide in the EU has been validated by the European Medicines Agency (EMA) and is currently under review for the treatment of adult patients with relapsed or refractory DLBCL, including DLBCL arising from low grade lymphoma, who are not candidates for ASCT.

Tafasitamab is being clinically investigated as a therapeutic option in B-cell malignancies in a number of ongoing combination trials.

Monjuvi is a registered trademark of MorphoSys AG.

XmAb is a registered trademark of Xencor, Inc.

Important Safety Information

What are the possible side effects of MONJUVI?

MONJUVI may cause serious side effects, including:

The most common side effects of MONJUVI include:

These are not all the possible side effects of MONJUVI.

Call your doctor for medical advice about side effects. You may report side effects to FDA at 1-800-FDA-1088.

Before you receive MONJUVI, tell your healthcare provider about all your medical conditions, including if you:

You should also read the lenalidomide Medication Guide for important information about pregnancy, contraception, and blood and sperm donation.

Tell your healthcare provider about all the medications you take, including prescription and over-the-counter medicines, vitamins, and herbal supplements.

Please see the full Prescribing Information for Monjuvi, including Patient Information, for additional Important Safety Information.

About Incyte

Incyte is a Wilmington, Delaware-based, global biopharmaceutical company focused on finding solutions for serious unmet medical needs through the discovery, development and commercialization of proprietary therapeutics. For additional information on Incyte, please visit Incyte.com and follow @Incyte.

About MorphoSys

MorphoSys (FSE & NASDAQ: MOR) is a commercial-stage biopharmaceutical company dedicated to the discovery, development and commercialization of innovative therapies for patients suffering from cancer and autoimmune diseases. Based on its leading expertise in antibody, protein and peptide technologies, MorphoSys, together with its partners, has developed and contributed to the development of more than 100 product candidates, of which 27 are currently in clinical development. In 2017, Tremfya, developed by Janssen Research & Development, LLC and marketed by Janssen Biotech, Inc., for the treatment of plaque psoriasis, became the first drug based on MorphoSys antibody technology to receive regulatory approval. In July 2020, the U.S. Food and Drug Administration (FDA) granted accelerated approval of MorphoSys proprietary product Monjuvi (tafasitamab-cxix) in combination with lenalidomide in patients with a certain type of lymphoma. Headquartered near Munich, Germany, the MorphoSys group, including the fully owned U.S. subsidiary MorphoSys US Inc., has more than 600 employees. More information at http://www.morphosys.com or http://www.morphosys-us.com.

Monjuvi is a registered trademark of MorphoSys AG.

Tremfya is a registered trademark of Janssen Biotech, Inc.

Incyte Forward-Looking Statements

Except for the historical information set forth herein, the matters set forth in this press release, including statements regarding Incytes ongoing clinical development program for tafasitamab in patients with relapsed/refractory follicular lymphoma (FL) or marginal zone lymphoma (MZL), the enrollment, design, and timing and results of the clinical trial program, including the inMIND study, and whether tafasitamab will become an approved treatment option for patients with relapsed or refractory FL or relapsed or refractory MZL, contain predictions, estimates and other forward-looking statements.

These forward-looking statements are based on Incytes current expectations and subject to risks and uncertainties that may cause actual results to differ materially, including unanticipated developments in and risks related to: unanticipated delays; further research and development and the results of clinical trials possibly being unsuccessful or insufficient to meet applicable regulatory standards or warrant continued development; the ability to enroll sufficient numbers of subjects in clinical trials; determinations made by the FDA; Incytes dependence on its relationships with its collaboration partners; the efficacy or safety of Incytes products and the products of the Incytes collaboration partners; the acceptance of Incytes products and the products of Incytes collaboration partners in the marketplace; market competition; sales, marketing, manufacturing and distribution requirements; greater than expected expenses; expenses relating to litigation or strategic activities; and other risks detailed from time to time in Incytes reports filed with the Securities and Exchange Commission, including its Form 10-K for the year ended December 31, 2020. Incyte disclaims any intent or obligation to update these forward-looking statements.

MorphoSys Forward-Looking Statements

This communication contains certain forward-looking statements concerning the MorphoSys group of companies, including the expectations regarding Monjuvi's ability to treat patients with relapsed or refractory diffuse large B-cell lymphoma, the further clinical development of tafasitamab-cxix, including ongoing confirmatory trials, additional interactions with regulatory authorities and expectations regarding future regulatory filings and possible additional approvals for tafasitamab-cxix as well as the commercial performance of Monjuvi. The words "anticipate," "believe," "estimate," "expect," "intend," "may," "plan," "predict," "project," "would," "could," "potential," "possible," "hope" and similar expressions are intended to identify forward-looking statements, although not all forward-looking statements contain these identifying words. The forward-looking statements contained herein represent the judgment of MorphoSys as of the date of this release and involve known and unknown risks and uncertainties, which might cause the actual results, financial condition and liquidity, performance or achievements of MorphoSys, or industry results, to be materially different from any historic or future results, financial conditions and liquidity, performance or achievements expressed or implied by such forward-looking statements. In addition, even if MorphoSys' results, performance, financial condition and liquidity, and the development of the industry in which it operates are consistent with such forward-looking statements, they may not be predictive of results or developments in future periods. Among the factors that may result in differences are MorphoSys' expectations regarding risks and uncertainties related to the impact of the COVID-19 pandemic to MorphoSys' business, operations, strategy, goals and anticipated milestones, including its ongoing and planned research activities, ability to conduct ongoing and planned clinical trials, clinical supply of current or future drug candidates, commercial supply of current or future approved products, and launching, marketing and selling current or future approved products, the global collaboration and license agreement for tafasitamab, the further clinical development of tafasitamab, including ongoing confirmatory trials, and MorphoSys' ability to obtain and maintain requisite regulatory approvals and to enroll patients in its planned clinical trials, additional interactions with regulatory authorities and expectations regarding future regulatory filings and possible additional approvals for tafasitamab-cxix as well as the commercial performance of Monjuvi, MorphoSys' reliance on collaborations with third parties, estimating the commercial potential of its development programs and other risks indicated in the risk factors included in MorphoSys' Annual Report on Form 20-F and other filings with the U.S. Securities and Exchange Commission. Given these uncertainties, the reader is advised not to place any undue reliance on such forward-looking statements. These forward-looking statements speak only as of the date of publication of this document. MorphoSys expressly disclaims any obligation to update any such forward-looking statements in this document to reflect any change in its expectations with regard thereto or any change in events, conditions or circumstances on which any such statement is based or that may affect the likelihood that actual results will differ from those set forth in the forward-looking statements, unless specifically required by law or regulation.

1 Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016;127:2375-2390.2 Decision Resources Group. Non-Hodgkins Lymphoma and Chronic Lymphocytic Leukemia, Landscape & Forecast. 2020.

View post:
Incyte and MorphoSys Announce First Patient Dosed in Phase 3 inMIND Study Evaluating the Addition of Tafasitamab to Lenalidomide and Rituximab in...

The role of stem cells in cancer progression and therapy | OTT – Dove Medical Press

Introduction

Conventionally, cancer therapy has relied on various pharmacological and radiation-based interventions, often through means of chemotherapy and radiotherapy.13 Current challenges in the clinical success of cancer therapy result from limitations in the interventional mechanisms themselves. Often, this is due to patient incompatibility with treatment, a unique disease phenotype or rapid drug resistance.4 This results in low rates of patient remission and greater rates of mortality.4 A new and developing area of research has opened up the realm of cancer therapy through a deeper focus on a novel interventional paradigm for cancer: stem cell therapy. Although stem cell therapy has remained an ongoing area of research with many new developments in cell-based therapies (CBT) for different diseases including autoimmune disorders and regenerative medicine, the molecular relationship between cancer stem cells (CSCs) and cancer pathogenesis has now grown into a budding realm of interest.5 This is due to various studies that have highlighted the critical role of CSCs in promoting a tumorigenic environment.6 A greater focus on researching the role of stem cells including CSCs in cancer progression and development will permit the creation of novel therapies and technologies that can target cancers at earlier stages of pathogenesis. This can also allow for long-term resolutions to many cancers because the highly transformative properties of stem cells can be repurposed for targeting cancer cells through genetic or phenotypic alteration, a mechanism which is in contrast to short-term remedies like chemotherapy.7 The current reliance on radiation therapy for cancer and the use of chemo drugs which impact healthy, endogenous cellular functions results in greater, more lethal side effects.8,9 To circumnavigate this issue, a focus on genetic and molecular therapies that do not impact normal, healthy cell function can support the development of longitudinal therapies for cancer with reduced side effects and morbidity rates. This approach is made feasible through studying the interactions amongst components of the tumor microenvironment, particularly with a focus on stem cell interactions and cell-conversion in cancer pathogenesis. Hence, due to its potential to transform the current narrative in the approach to cancer therapy, here we explore the current state of the role of stem cells in cancer progression and therapy. Particularly, we summarize numerous studies that explore potential areas of focus when targeting stem cells for cancer therapy, including targeting markers on CSCs, stem cells for immunization against tumors, using stem cells as carriers of therapeutic cargo, quiescent properties of stem cells, and targeting exosomes to prevent (Epithelial Mesenchymal Transition) EMT and metastasis.

Primary cancer cells have been found to derive from stem cells, with a subsection of these cells named cancer stem cells (CSCs). CSCs replicate similar characteristics to regular stem cells, such as the ability to proliferate in their microenvironments.10 CSCs sustain the cancer by promoting proliferation, and therefore must be targeted when attempting to eliminate cancer for successful and long-lasting results.11 As do most healthy cells and hematopoietic stem cells, pancreatic, liver, and lung CSCs overexpress CD-47 on their cell surface, which is an immunosuppressive defense signal that inhibits attacks from macrophages on cancer cells.12 Normally, CD-47 plays a positive role of defense in protecting healthy cells in organs and tissue from harmful attack by macrophages.13,14 Similarly, in CSCs, the CD-47 protein acts as an immune checkpoint blockade for any targeted attempts to diminish the CSCs by activated macrophages in the tumor microenvironment.15,16 CD-47 elicits its effects via interaction with signal regulatory protein alpha (SIRP) to inhibit phagocytosis of normal cells, which suppresses the phagocytic activity of immune cells, particularly macrophage phagocytosis.17 CD-47 is overexpressed in many human malignancies.18 One study performed simultaneous silencing of CD-47 and PD-L1 in order to enhance immunotherapy against circulating tumor cells.19 Inhibiting PD-L1 allowed immune cells to locate tumor cells more adequately, and blockade of CD-47 permitted macrophage-mediated destruction of the tumor cells. In vitro flow cytometry confirmed overexpression of CD-47 and PD-L1 in the tumor cell line.19 Compared to the blank controls or single-antibody group, dual inhibition of these immunosuppressive proteins resulted in a more potent reduction of solid tumors in mice.

In order to target CD-47 and silence its downstream effects, various forms of pharmacological and nanomedicine-based approaches have been established. An antibody named Hu5F9-G4 that targets CD-47, allowing macrophages to destroy the cancer cells, has been developed. Another similar antibody, Rituximab, which has been known to positively amplify destruction signals inhibited by CD-47, is highly active and is well tolerated as first-line single-agent therapy for indolent non-Hodgkin lymphoma (NHL).20 Using the application of both antibodies, Hu5F9-G4 and Rituximab, the results of a clinically evaluated study on the treatment outcome in patients of NHL concluded that at least 50% of the test subjects had eliminated most symptoms of cancer.21 The authors did not report directly on tumor size, but clinical evaluation of response to treatment indicated that more than 60% of patients had complete or partial response to the drug, a metric that relates to change in tumor size. PET-CT of patients indicated partial to complete remission of the lymphoma in male and female adult subjects.21 This method of targeting CD-47 protein on Cancer Stem Cells has been shown to result in no detectable side effects in human beings, and thus may pave new routes for immunotherapy towards many forms of cancer by targeting cancer stem cells.22 This is because in the past, various CSCs in cancers such as pancreatic, lung, and breast cancers have been proven to express CD-47.2325 Hence, targeting this immune blockade molecule expressed on CSCs may provide a new avenue of cancer treatment.

However, current limitations to therapies targeting CD47 result from its presence on an abundance of normal, healthy cells. Thus, the attack on host cells as a side effect of treatment is a potential roadblock in CD47-based therapy. To circumnavigate this issue, several CSC relevant markers have been elucidated in efforts to target CSCs specifically without damaging native cells. CD44 and CD133 are widely used and accepted as relevant cell-surface markers for CSCs.2630 CD44 has implications in being a CSC marker for breast, pancreatic, and head and neck cancers.28 The enrichment of CD44 cells after drug treatment indicates higher rates of proliferation and a greater resistance to drug-induced death, helping dictate CD44 as a negative prognostic factor.31 CD133 holds implications in acting as a CSC marker for brain, colon, and prostate cancers.32 CD133 has been shown to upregulate the FLIP (FLICE-like inhibitory protein) which aids in CSC resistance to apoptosis.32 CD133 is also a vastly recognized chemo-resistant CSC surface antigen, thus aiding in the prognosis of treatment. By inhibiting CD133, the cells proliferation pathways are also inhibited. A larger presence of CD133 on the tumor may require alternative chemotherapeutic agents, or a different treatment.32

The benefit of identifying CSC-surface molecules lies in their potential to act as targets for new cytotoxic therapies,33 often those which are mediated by neutralizing antibodies, in combination with CSC-directed therapy. Several studies have recently explored this form of combined therapy which prevents the repeated formation of tumors and inhibits recurrent population of CSCs. One such study evaluated the effect of a combined therapy on drug-resistant triple-negative breast cancer (TNBC) cells by targeting the TGF- of the CSC.34 Particularly, the group demonstrated that the TNBC CSC population maintained a unique ability to upregulate IL-8 in response to TGF- signaling following chemotherapy with Paclitaxel (a mechanism which contributes to drug resistance). Using a TGF- type 1 receptor kinase inhibitor, a TGF- type II receptor neutralizing antibody, and SMAD-4 siRNA as forms of combined therapy with Paclitaxel, the subsequent recruitment of IL-8 following chemotherapy was blocked and the expansion of the chemotherapy-resistant CSC populations was inhibited. This study shows the ability of combined therapy to both inhibit primary mammosphere and further prevent drug resistance in the CSC population of the TNBC population. Besides above-mentioned markers, Aldehyde dehydrogenase 1 (ALDH1) has also been reported as a marker of cancer stem cells could be targeted for molecular therapy.35,36 Table 1 provides a list of common cell-surface and intracellular markers pertaining more specifically to CSCs.

Table 1 A List of Common Cell-Surface and Intracellular Markers Pertaining to CSCs

At the beginning of the 20th century, Frederick Schne noted that fetal tissue vaccination could suppress transplanted tumor growth in mice.37 However, it took many more years for other groups to further investigate the potential of this discovery. In the 1960s and 70s, research in this area resumed, and investigators reported mice immunized against embryonic material could prevent tumor growth, priming their bodies to recognize and fight cancer cells. However, these results tended to be weak and hard to reproduce.3840 Furthermore, ethical concerns and technological limitations during this time period made further research in humans impossible. With recent progress involving embryonic cell lines, research into this area has been revisited. These include studies that found very similar RNA transcript profiles and surface antigen expression between embryonic cells and different cancer cell lines, including pancreatic cancer, prostate cancer, breast cancer, myeloid leukemia, and glioblastoma.41,42 Furthermore, ES and cancer cells have both been shown to exhibit similar markers of stemness, particularly when these cancer cells are less differentiated, or more immature.42 In this study by Ben-Porath et al, poorly differentiated breast tumors were shown to display an ESC-like expression signature, more so than further differentiated tumors. As ESCs are known to exhibit stemness, this shows the possibility that more immature tumors (less differentiated) may exhibit higher tumor stemness than mature tumors. On this point of stemness, stemness is usually described as the ability for stem cells to balance between a few different processes: proliferation, quiescence, regeneration, and differentiation.43 Stem cells rely on interactions with and signals within their microenvironment to determine which of these processes to undergo.44 Cancer cells exhibit stemness as well, and can use this stemness to survive stress and treatment, and preserve their lineage.43 Discovering the similarity in gene and surface antigen expression between embryonic stem cells (ESCs) and cancer cells, we now understood why these cells could potentially be used to as an anti-tumor vaccination. Moreover, since the discovery of induced pluripotent stem cells, groups have shown that the transcriptomes and antigens of ESCs and iPSCs are almost identical,45 and that induced pluripotent stem cells (iPSCs) also show potential as an immunization agent. In fact, iPSC is most likely advantageous compared to ESCs for this purpose, as using iPSCs from a specific patient would be more representative of the patients own immunogens.46 Furthermore, a theoretically unlimited number of iPSCs can be generated from each patient, given enough time. In 2018, a study used tumor-specific antigens and tumor-associated antigens expressing iPSCs to prime the immune systems of mice, followed by the transplantation of different tumors, including melanoma and breast cancer.47 Significant regression of these tumors was found when compared to the control group. This result was attributed to the upregulation in mature antigen-presenting cells in the lymph nodes, which led to an increase in helper and cytotoxic T-cells. This group then isolated T-cells and tumor-experienced lymphocytes (TELs) from mice with tumor and that had received vaccination and transferred them to mice with tumor that had not received vaccination. Both of these groups experienced tumor regression. However, it is important to note that this iPSC vaccination was only able to slow or prevent the growth of tumors that were transplanted after vaccination; it was not effective in preventing the growth of tumors that have already been established in vivo. This continues to be the shortcoming of using stem cells as a therapeutic treatment; their effect is diminished when tumor transplant and growth takes place before the vaccination. For this reason, they tend to provide better results when used as a prophylactic treatment, as opposed to a therapeutic treatment after disease onset. The safe application of ESC and iPSCbased technologies requires the use of methods of iPSCs production and their directed differentiation which minimize both the possibility of mutations in cell genomes under in vitro culturing and the probability of malignant transformation of the injected cells.48 Some have suggested that vaccinations including CSC lysates would improve outcomes, such as a vaccination that Lin et al formulated.49,50 They combined CSC dendritic cells, which present tumor-associated antigens to T cells, with melanoma and carcinoma tumor models, which showed promise in increasing protective immunity against tumor cell challenge. However, as pointed out in a recent review by Chu et al, the isolation of enough number of CSCs (in this case, CSC-DCs) from tumor tissues is very challenging, which poses difficulties in access and quantity for possible future study in larger animal models, or in the clinic.51 As discussed earlier, iPSCs can be made in large quantities and from each individual patient, but they do not provide adequate tumor suppression when tumor has been established. Combination vaccines with CSCs and tumor cells show promise but provide isolation and collection challenges. A very recent study, published in 2020, provides improvement on cancer vaccines for tumor rejection.52 In this study, this group enriched a whole-cell melanoma vaccine with stem cells (this vaccine also contained a molecular adjuvant, cytokine Hyper-IL6). One vaccine was enriched with melanoma stem-like cells from B16F10 melanospheres, while the other vaccine contained mouse-induced pluripotent stem cells (miPSCs). While both vaccines showed impressive reductions in tumor growth, and in disease-free and overall survival of the immunized mice, the most effective vaccine was the one containing miPSCs. This study provides exciting evidence that vaccines containing iPSCs with tumor cells can be just as effective if not more effective than those containing the harder to obtain tumor stem cells. However, once again, this study was only done with immunization before tumor transplant, so no conclusions about treatment after transplant and tumor establishment can be made. Currently, we can only suggest stem cell vaccine for cancer treatment in addition to other treatments such as surgery, radiation, and chemotherapy, and not as a standalone therapy option, as it has only been seen as effective before tumor formation and progression.47,51,53

Genetic modification enhances the therapeutic potential for stem cells in oncology by facilitating precise secretion of bioactive mediators. Typically derived from bone marrow, endogenous mesenchymal stem cells (MSCs) migrate towards sites of damaged tissue. MSC tropism is propagated by a cascade of signaling mechanisms and chemokines which trigger the recruitment of MSCs towards sites of damaged tissue.54,55 MSCs are able to mobilize effectively as they express numerous chemokine receptors including: CCR1, CCR2, CCR4, CCR7, CCR8, CCR9, CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, and CX3CR1.56 Additionally, MSCs possess the ability to produce a diverse array of cell adhesion molecules which facilitate the engraftment to specific target tissue.57 Upon transplantation, MSCs will migrate away from the initial injection site into a tumor microenvironment (TME) before engrafting to various target cells. Therefore, the transduction of MSCs and other multipotent stem cells could potentially facilitate the decisive delivery of a therapeutic payload within a tumor microenvironment. Specifically, virally transduced MSCs and Neural Stem Cells (NSCs) have exhibited the expression of chemotactic cytokines, interleukins, interferons, growth factors and prodrug-converting enzymes.58,59 The latter of which constitutes the technique known as gene-directed enzyme prodrug therapy (GDEPT). This treatment method allows various non-toxic prodrugs to be converted into their active forms via non-endogenous enzymes produced by genetically modified stem cells.60 The aggregate of these characteristics makes GDEPT uniquely qualified to treat gliomas, medulloblastomas, and other brain tumors. Another benefit of this therapy stems from the ability of MSCs to manipulate tight junctions within the blood brain barrier (BBB), temporarily inhibiting its exclusion properties and allowing for the seamless traversal of MSCs into the cortex; MSCs then utilize tumor-tropism mechanisms to infiltrate and destroy tumor cells in the brain.59,61

Another means by which stem cells can serve as therapeutic carriers is by the precise delivery of nanoparticles (NPs) bearing anti-cancer drugs and various other oncolytic mediators. NPs have long been used in the distribution of drugs used to treat cancer. However, the applicability of NPs is limited due to the lack of accurate targeting, their tendency to be internalized by a wide variety of normally function cells, and their rapid excretion from the body.62 One study analyzing the nanodrug deposits provided by MSCs internalized within mice found that NPs exhibited more accurate delivery of therapeutics in a developed orthotopic lung tumor.63 An additional study conducted using rats has demonstrated that MSCs infiltrate tumor tissue uniformly and that this infiltration leads to a more uniform distribution of a therapeutic payload. However, in the same study, they found no evidence to suggest MSCs could engage in long-distance tropism for a series of gliomas.64 Despite this, NPs conjugated to anti-cancer agents can be delivered into a tumor microenvironment reliably using stem cell-mediated tumor tropic delivery. Furthermore, MSCs retain their inherent ability to sense tumors and respond to chemokines following the anchoring of nanoparticles to their surface. In fact, there is no significant difference in tumor tropism between traditional MSCs and those bound to NPs.65 However, in the latter case, the half-life of the nanoparticle is increased exponentially.66,67

MSCs, with their innate tumor tropism characteristics, are ideal agents for this style of theranostic-based therapy. This is because the inherent tumor tropism presents various biomarkers that can be used as targets for nanoparticles which can then be imaged in vivo using clinically relevant imaging modalities such as Magnetic Resonance Imaging (MRI) and Nuclear imaging.53,6870 Nouri et al demonstrate that theranostic MSCs are a reliable cell-based, non-viral or viral vectors for suicide gene therapy of cancer using enzyme/prodrug systems.71 MSCs were used as a medium to perform the first comparative study that illustrated the impact of subtle differences among various enzyme/prodrug systems such as thymidine kinase/ganciclovir (TK/GCV), yeast cytosine deaminase/5-fluorocytosine (yCD/5-FC) and nitroreductase/CB1954 (NTR/CB1954) on the therapeutic outcome. MSCs were genetically modified to stably express a panel of four suicide genes including TK (TK007 and TKSR39 mutants), yeast cytosine deaminase: uracil phosphoribosyltransferase (yCD:UPRT) and nitroreductase (NTR). Then, they evaluated the anticancer efficacies of the genetically engineered MSCs using SKOV3 cell models in vivo. In addition, all MSCs were engineered to stably express luciferase gene making them suitable for quantitative imaging and doseresponse relationship studies in vivo. The study results demonstrated that yCD:UPRT/5-FC was the most effective enzyme/prodrug system among the ones tested with this theranostic imaging platform.71

It is worth noting that theranostics can be applied to nanodrug therapy for cancer. Liu et al reported that a silica-based multifunctional NP system encapsulated a chemotherapeutic agent and magnetic cores and coated with a specific antibody against the lung CSCs was systematically studied in vivo. These NPs were systematically administered and activated for targeted chemotherapy and thermotherapy by using an externally applied alternating magnetic field (AMF).72 The application of an AMF causes localized induced hyperthermia in the areas in which the nanoparticles accumulate.73 This can kill the tumor cells either directly or indirectly. The cytotoxic thermal effects of the localized hyperthermia from the NPs directly ablate the tumor cells. There is also an indirect mechanism which supports dendritic cell activation and immune cell migration to the tumor microenvironment. This is caused by the hyperthermia which induces the production of Heat Shock Protein (HSP) and a subsequent increase in presentation of these antigens on the cell surface, which induces dendritic cells and triggers a greater host immune response towards the cancer cells.74 The antibody-modified NPs targeted to lung CSCs with extended accumulation in tumors after systemic injection. In in vivo models, this hyperthermia and chemotherapeutic combined therapy significantly suppressed tumor growth and metastasis in lung CSC xenograft-bearing mice, with minimal side effects and adverse effects.72 This work demonstrated the feasibility of developing multifunctional nanomedicine targeting CSCs for effective cancer treatment, which can be monitored by magnetic particle imaging or MRI.7577 Figure 1 highlights some of the aforementioned mechanisms of using stem cells as carriers of therapeutic payload and alternative therapeutic carriers such as NPs for tumor treatment.

Figure 1 Therapeutic carrier models for cancer treatment. (Left) Cancer stem cells (CSCs) loaded with therapeutic cargo can be transplanted or delivered to site of tumor formation (tumor bed). (Right) Nanoparticles (NPs) targeting the CSCs can deliver chemotherapeutic payload and induce hyperthermia with an external applied magnetic field (AMF).

Anti-cancer therapies often utilize a combination of procedures and drugs that target gatherings of tumor cells. CSCs have long been understood now as a group of cells that fuel the growth of tumors and have properties that allow them to persist through the electromagnetic and chemical treatment that are common in contemporary practice.78 CSCs separate themselves among cancer cells due to their ability to maintain a long, slow growing quiescent state. This dormancy allows for the cells to be preserved in spite of conventional cancer treatments that are used to combat their progeny as well as posing long-term tumorigenic potential.78 Previous studies have shown that there maintains a population of chemotherapy-resistant cancer cells that demonstrated unique properties of self-renewal and increased potential for tumor formation. In these studies, CSC-marker expressing cells have survived treatment such as neoadjuvant chemotherapy whereas cells without CSC markers were destroyed.79

Their unique properties have only added on to the need for the development of new therapeutic strategies that exceed the scope of conventional antiproliferative agents and treatments. Much research is focused on CSCs quiescent properties as a potential target for treatment. Research has shown that this quiescent function is not simply a dormant state, but rather it is actively maintained by the cell by downregulating known regulators of the cell cycle such as cyclin A2 and E2 as well as mitotic regulators such as survivin.80 There are generally three main approaches through which research is attempting to eradicate these quiescent stem cells in tumors. The first involves driving quiescent cells to reenter a normal cell cycle state by stopping cellular mechanisms that drive quiescence and then attack them with chemotherapeutic agents that only function on proliferating cells.81 This theory is primarily supported by the notion that cell quiescence is defined as being in a reversible G0 state that requires maintenance.81 For example, Fbxw7 has been understood to play a crucial role in the maintenance of quiescence, but its ablation along with the introduction of Imatinib treatment, a tyrosine-kinase inhibitor, is shown to interrupt the quiescent state of the studied leukemia-initiating cells (LICs) and contribute to their depletion.82 The second method that has been proposed is one where pharmacological intervention is used to maintain CSCs in their quiescent state throughout the lifetime of their patient so that they will not cause any future tumor growth or metastasis. This strategy attempts to lock these quiescent cells in the G0 state. One such way to accomplish this is to inhibit Src kinase signaling along with inhibiting MEK1/2 as they are both factors in cell cycle progression. This can be achieved through pharmacological means and the prevention of cell cycle progression will induce apoptosis or maintenance in this state without growth.83 The final school of thought looks to eradicate CSCs while they are still in their dormant state.81 However, there are issues with these three potential methods which further highlight why research in these fields has been unsuccessful. Clinical evidence has been inconclusive on whether activating these quiescent cells in order to target them with conventional therapy is even able to be controlled. In addition, these quiescent stem cells are known to be extremely heterogeneous which indicates that activating them could exacerbate the condition by giving them an increased arsenal of mechanisms through which the cancer can develop.84 Difficulties in the latter two methods are due to the need of a more comprehensive understanding of the various pathways and factors associated in CSC quiescence. Normal stem cells are a valuable resource in understanding CSC signaling as they are known to share several pathways such as Notch, Hedgehog, WNT/B-catenin, and NFkB.81,85

Multiple myeloma (MM) is a common cancer of the blood which has an alarming 5-year survival rate of about 54% along with high chances of recurrence and need for further treatment.86 Its resistance to treatments has been attributed to its development of drug resistance. A proposed model of MM malignant stem cell progression suggests that there are two states that cells are in - a quiescent state and a proliferative state. The quiescent state has exhibit properties of increased adhesion and minimal proliferation. Those in the quiescent state also have increased drug-resistant properties. There also exists a small population of cells that are in the proliferative state. These cells have high capacity for growth and mutation as a result of their increased chromosomal instability.87 Impairment of MM cell survival through inducing cell cycle arrest was conducted through the use of anti-sense oligonucleotides (ASOs) to target human interferon regulatory factor 4 (IRF4). IRF4 has been identified as a critical MM cell factor for survival with an important role in disease development and progression. This treatment also reduced mRNA levels and levels of the MYC gene, which has a known impact in stem cell progression and has been studied thoroughly as an oncogene. Increased cleavage rates, apoptosis rates, and decreased colony formation all suggested success in disrupting cell growth. Furthermore, ASO-mediated IRF4 treatment led to cell cycle arrest in G1 and decreased proportion of cells in G2/M phase which signifies significant reduction in cell viability.86 Seeing as though the G1 phase length plays an important factor in maintaining a quiescent state, this ASO treatment could help decrease the risk of quiescent MM cells leading to disease relapse. Further study is necessary to find an effective solution to both quiescent and proliferative states of MM, but studies that combat CSC quiescence show promise for a solution to decrease relapse rates.

Much of the difficulty in identifying, understanding, and treating CSCs originates from their extensive plasticity and asymmetry driven by intra-tumoral heterogeneity.34 Their plasticity allows them to evade therapies by presenting various phenotypes and providing the ability to inhabit different tumor microenvironments. Normal cancer cells within a tumor mass can convert into CSCs in response to chemotherapy and can gain drug resistance as a result of a change in gene expression.88 One such mechanism of drug resistance includes an increase in the expression of ATP-Binding cassette (ABC) transporters that permit greater drug efflux rates. CSCs can also be found in either a quiescent state that is difficult to eradicate or a proliferative state that prioritizes growth and metastasis. This quiescent state, when met with increased drug efflux mechanisms in CSC populations, often contributes to greater relapsed tumors. In hypoxic conditions, cells seeded in the core of the tumor are found to transition to the edge of the tumor where they become more quiescent.89 This is because cells typically found seeded in the tumor core are more proliferative, which is why there is greater cell density in that region as compared to the edge cells which are more invasive, quiescent, and resistant.89 As a result of this core-to-edge migration of tumor cells in response to hypoxia, particularly due to the induction of HIF-1 and HIF-2, a novel front for cancer intervention and therapy has been unlocked.89 However, the therapeutic potential and efficacy of a combined therapy targeting HIF-1 and HIF-2 in tumor cells, likely to prevent core to edge migration of progressive tumoral cells when presented with hypoxia, needs to be further studied to gain momentum as an effective therapy. The cell division of these quiescent cells can also be either symmetric or asymmetric. Symmetric division either creates a pair of quiescent daughter cells or a pair that is more proliferative and differentiates.90 Asymmetric division results in one quiescent cell and transient amplifying cell that is very proliferative and contributes to most of the tumor.90 It is widely inferred that this asymmetric division is more dangerous for cancer growth and it should be targeted for effective cancer treatment.

Stem cells have been found to play the role of mediators of conversion of healthy cells to cancerous cells.91 This is done through the synthesis and release of exosomes extracellular vesicles of cellular cargo that are released by a cell for various purposes.92 Exosomes have been previously found to contain various cargo including miRNA, siRNA, transcription factors, and other proteins. Through the use of these extracellular vesicles, cells are able to communicate and exert transformative influence through phenotypic and genotypic alterations of surrounding cells.93 Often used to encourage differentiation, proliferation, or inhibition thereof of cells in the surrounding environment, exosomes role in mediating the transformation of regular, healthy cells into cancerous cells is an area of great research due to its potential to catalyze interventional mechanisms for cancer therapy through targeting exosomes.

Stem cell-derived extracellular vesicles can provide powerful alternatives to cell-based therapies since the former acts as a noninvasive method for in vivo modulation of gene expression, inhibition of cell surface receptors and intracellular signaling molecules, and initiation of cell differentiation or death.92 Instead of transplanting differentiated stem cells or donor cells into the patient, the delivery of extracellular vesicles can allow for transformation of native cells in the subject and transformation of cells that have been transplanted prior. This eliminates the need for surgery and removes the necessity to probe the patient for cell transplantation, which requires some invasive protocol. Stem cell-derived extracellular vesicles can prove to be a valuable tool in cell-based therapy by perhaps altering the nature of the cells that are native to the microenvironment of the subject.

There are various mechanisms through which exosomes released by CSCs exert influence on neighboring cells. One such method is through induction of Epithelial to Mesenchymal Transition (EMT). This is induced through the release of transcription factors such as Snail, Twist, and FOXC2 that are carried by the extracellular vesicle to neighboring cells where these transcription factors exert their effects phenotypically and genotypically.94 In mammary epithelial stem cells, the influence of these factors on neighboring healthy cells is apparent by the CD44 (high)/CD24 (low) antigen phenotype which is typical of CSCs.95 This can contribute to the development of severe tumorigenicity amongst the cells, as noted by the study. Another study observed the role of CD-103+exosomes released by CSCs which promoted EMT in clear cell renal cell carcinoma (CCRCC) cells.96 Particularly, the group observed the importance of one critical factor in promoting EMT in CCRCC: MiR-19b-3p.96 This was done through the repression of PTEN, a protein which is apparently expressed and has been previously studied to play a key role in the promotion of EMT.97 The study was able to confirm the role of MiR-19b-3p in promoting EMT through suppression of PTEN by infecting ACHN and 786-O cells with a miR-19b-3p lentivirus.96 Like the exosomes released by the CSCs, this encouraged the migration and invasion of the CCRCC cells. These findings implicate the role of CD103, since it mediates entry of the miRNA into neighboring cells via use of exosomes, in acting as a potential biomarker or target for cancer therapy. Further studies focusing on CSC-derived exosomes include macrovesicles derived from CD105+ cells which have been found to promote angiogenesis and metastasis with a distinct miRNA profile inside of the vesicles, resulting from presence of CD105.98 This unique composition encouraged greater tumorigenicity amongst the cells, favoring further growth and invasion. Although there are various studies investigating the role of CSC-derived exosomes in promoting tumor development, limited information currently exists on the CSC-specificity of these exosomes and further research must be done in this area to determine entirely which exosomes are specific to only CSCs, or whether it is the content within that can be used as a biomarker. This further emphasizes the importance of researching the role of the CSC-influenced tumor microenvironment and its various components (eg, exosomes) in promoting and accelerating cancer development.

The aforementioned studies permit insight into a previously unexplored interface between stem cells and cancer progression and treatment. Silencing cell surface markers on CSCs can promote immune recognition of tumor sites and inhibit binding of tumor cargo to healthy cells, thus preventing their transformation. Furthermore, iPSCs can be used to vaccinate and immunize individuals against tumor cells in the future due to surface-antigen similarity between iPSCs and cancer cells. In addition to these mechanisms, stem cells have been shown to transfer various genetic and molecular cargo intercellularly through exosomes, another area for drug targeting and disease intervention in cancer. As a result of these various interactions between cancer cells, stem cells and their individual components, there is now a greater need to explore the influence that stem cells have on tumorigenesis. This will enable the innovation and translation of theranostics that are more efficient in nature and do not result in an abundance of unwanted side effects as a result of treatment.

Current organoid models are limited by their inability to mimic mature organ architecture and associated tissue microenvironments. Multilayer bladder assembloids were recently created by reconstituting tissue stem cells with stromal components to represent an organized architecture with an epithelium surrounding stroma and an outer muscle layer.99 A urothelial carcinoma assembloids platform has been developed by the same group. These assembloids exhibit characteristics of mature adult bladders and tumors in cell composition and gene expression, and recapitulate in vivo tissue dynamics. This will help advance functional studies in the context of the increasingly recognized importance of tissue stroma and microenvironments.99101

The authors report no conflicts of interest in this work.

1. Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol. 2014;740:364378. doi:10.1016/j.ejphar.2014.07.025

2. Montero A, Fossella F, Hortobagyi G, Valero V. Docetaxel for treatment of solid tumours: a systematic review of clinical data. Lancet Oncol. 2005;6(4):229239. doi:10.1016/S1470-2045(05)70094-2

3. Kim N, Choi SH, Chang JS, et al. Use of bevacizumab before or after radiotherapy increases the risk of fistula formation in patients with cervical cancer. Int J Gynecol Cancer. 2020;31:5965. doi:10.1136/ijgc-2020-002031

4. Lage H. An overview of cancer multidrug resistance: a still unsolved problem. Cell Mol Life Sci. 2008;65(20):31453167. doi:10.1007/s00018-008-8111-5

5. Biehl JK, Russell B. Introduction to stem cell therapy. J Cardiovasc Nurs. 2009;24(2):98. doi:10.1097/JCN.0b013e318197a6a5

6. Ayob AZ, Ramasamy TS. Cancer stem cells as key drivers of tumour progression. J Biomed Sci. 2018;25(1):20. doi:10.1186/s12929-018-0426-4

7. Zhang CL, Huang T, Wu BL, He WX, Liu D. Stem cells in cancer therapy: opportunities and challenges. Oncotarget. 2017;8(43):7575675766. doi:10.18632/oncotarget.20798

8. Garibaldi C, Jereczek-Fossa BA, Marvaso G, et al. Recent advances in radiation oncology. Ecancermedicalscience. 2017;11:785. doi:10.3332/ecancer.2017.785

9. Urruticoechea A, Alemany R, Balart J, Villanueva A, Vinals F, Capella G. Recent advances in cancer therapy: an overview. Curr Pharm Des. 2010;16(1):310. doi:10.2174/138161210789941847

10. Brown EJ, Frazier WA. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 2001;11(3):130135. doi:10.1016/S0962-8924(00)01906-1

11. Blazar BR, Lindberg FP, Ingulli E, et al. CD47 (integrin-associated protein) engagement of dendritic cell and macrophage counterreceptors is required to prevent the clearance of donor lymphohematopoietic cells. J Exp Med. 2001;194(4):541549. doi:10.1084/jem.194.4.541

12. Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD, Lindberg FP. Role of CD47 as a marker of self on red blood cells. Science. 2000;288(5473):20512054. doi:10.1126/science.288.5473.2051

13. Jaiswal S, Jamieson CH, Pang WW, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138(2):271285. doi:10.1016/j.cell.2009.05.046

14. Olsson M, Bruhns P, Frazier WA, Ravetch JV, Oldenborg PA. Platelet homeostasis is regulated by platelet expression of CD47 under normal conditions and in passive immune thrombocytopenia. Blood. 2005;105(9):35773582. doi:10.1182/blood-2004-08-2980

15. Oronsky B, Carter C, Reid T, Brinkhaus F, Knox SJ. Just eat it: a review of CD47 and SIRP-alpha antagonism. Semin Oncol. 2020;47(23):117124. doi:10.1053/j.seminoncol.2020.05.009

16. Uno S, Kinoshita Y, Azuma Y, et al. Antitumor activity of a monoclonal antibody against CD47 in xenograft models of human leukemia. Oncol Rep. 2007;17(5):11891194.

17. Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138(2):286299. doi:10.1016/j.cell.2009.05.045

18. Wang CL, Lin MJ, Hsu CY, et al. CD47 promotes cell growth and motility in epithelial ovarian cancer. Biomed Pharmacother. 2019;119:109105. doi:10.1016/j.biopha.2019.109105

19. Lian S, Xie R, Ye Y, et al. Simultaneous blocking of CD47 and PD-L1 increases innate and adaptive cancer immune responses and cytokine release. EBioMedicine. 2019;42:281295. doi:10.1016/j.ebiom.2019.03.018

20. Hainsworth JD, Litchy S, Burris HA, et al. Rituximab as first-line and maintenance therapy for patients with indolent non-hodgkins lymphoma. J Clin Oncol. 2002;20(20):42614267. doi:10.1200/JCO.2002.08.674

21. Advani R, Flinn I, Popplewell L, et al. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkins lymphoma. N Engl J Med. 2018;379(18):17111721. doi:10.1056/NEJMoa1807315

22. Eladl E, Tremblay-LeMay R, Rastgoo N, et al. Role of CD47 in hematological malignancies. J Hematol Oncol. 2020;13(1):96. doi:10.1186/s13045-020-00930-1

23. Cioffi M, Trabulo S, Hidalgo M, et al. Inhibition of CD47 effectively targets pancreatic cancer stem cells via dual mechanisms. Clin Cancer Res. 2015;21(10):23252337. doi:10.1158/1078-0432.CCR-14-1399

24. Kaur S, Elkahloun AG, Singh SP, et al. A function-blocking CD47 antibody suppresses stem cell and EGF signaling in triple-negative breast cancer. Oncotarget. 2016;7(9):1013310152. doi:10.18632/oncotarget.7100

25. Liu L, Zhang L, Yang L, et al. Anti-CD47 antibody as a targeted therapeutic agent for human lung cancer and cancer stem cells. Front Immunol. 2017;8:404. doi:10.3389/fimmu.2017.00404

26. Dietrich A, Tanczos E, Vanscheidt W, Schopf E, Simon JC. High CD44 surface expression on primary tumours of malignant melanoma correlates with increased metastatic risk and reduced survival. Eur J Cancer. 1997;33(6):926930. doi:10.1016/S0959-8049(96)00512-6

27. Thapa R, Wilson GD. The importance of CD44 as a stem cell biomarker and therapeutic target in cancer. Stem Cells Int. 2016;2016:2087204. doi:10.1155/2016/2087204

28. Ranji P, Salmani Kesejini T, Saeedikhoo S, Alizadeh AM. Targeting cancer stem cell-specific markers and/or associated signaling pathways for overcoming cancer drug resistance. Tumour Biol. 2016;37(10):1305913075. doi:10.1007/s13277-016-5294-5

29. Shmelkov SV, Butler JM, Hooper AT, et al. CD133 expression is not restricted to stem cells, and both CD133+ and CD133- metastatic colon cancer cells initiate tumors. J Clin Invest. 2008;118(6):21112120. doi:10.1172/JCI34401

30. Brugnoli F, Grassilli S, Al-Qassab Y, Capitani S, Bertagnolo V. CD133 in breast cancer cells: more than a stem cell marker. J Oncol. 2019;2019:7512632. doi:10.1155/2019/7512632

31. Yoshida K, Tsujimoto H, Matsumura K, et al. CD47 is an adverse prognostic factor and a therapeutic target in gastric cancer. Cancer Med. 2015;4(9):13221333. doi:10.1002/cam4.478

32. Barzegar Behrooz A, Syahir A, Ahmad S. CD133: beyond a cancer stem cell biomarker. J Drug Target. 2019;27(3):257269. doi:10.1080/1061186X.2018.1479756

33. Bellomo C, Caja L, Moustakas A. Transforming growth factor beta as regulator of cancer stemness and metastasis. Br J Cancer. 2016;115(7):761769. doi:10.1038/bjc.2016.255

34. Bhola NE, Balko JM, Dugger TC, et al. TGF-beta inhibition enhances chemotherapy action against triple-negative breast cancer. J Clin Invest. 2013;123(3):13481358. doi:10.1172/JCI65416

35. Rabinovich I, Sebastiao APM, Lima RS, et al. Cancer stem cell markers ALDH1 and CD44+/CD24- phenotype and their prognosis impact in invasive ductal carcinoma. Eur J Histochem. 2018;62(3). doi:10.4081/ejh.2018.2943.

36. Tomita H, Tanaka K, Tanaka T, Hara A. Aldehyde dehydrogenase 1A1 in stem cells and cancer. Oncotarget. 2016;7(10):1101811032. doi:10.18632/oncotarget.6920

37. Brewer BG, Mitchell RA, Harandi A, Eaton JW. Embryonic vaccines against cancer: an early history. Exp Mol Pathol. 2009;86(3):192197. doi:10.1016/j.yexmp.2008.12.002

38. Buttle GA, Frayn A. Effect of previous injection of homologous embryonic tissue on the growth of certain transplantable mouse tumours. Nature. 1967;215(5109):14951497. doi:10.1038/2151495a0

39. Klavins JV, Mesa-Tejada R, Weiss M. Human carcinoma antigens cross reacting with anti-embryonic antibodies. Nat N Biol. 1971;234(48):153154. doi:10.1038/newbio234153a0

40. Yaddanapudi K, Mitchell RA, Putty K, et al. Vaccination with embryonic stem cells protects against lung cancer: is a broad-spectrum prophylactic vaccine against cancer possible? PLoS One. 2012;7(7):e42289. doi:10.1371/journal.pone.0042289

41. Ghosh Z, Huang M, Hu S, Wilson KD, Dey D, Wu JC. Dissecting the oncogenic and tumorigenic potential of differentiated human induced pluripotent stem cells and human embryonic stem cells. Cancer Res. 2011;71(14):50305039. doi:10.1158/0008-5472.CAN-10-4402

42. Ben-Porath I, Thomson MW, Carey VJ, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008;40(5):499507. doi:10.1038/ng.127

43. Aponte PM, Caicedo A. Stemness in cancer: stem cells, cancer stem cells, and their microenvironment. Stem Cells Int. 2017;2017:5619472. doi:10.1155/2017/5619472

44. Mushtaq M, Kovalevska L, Darekar S, et al. Cell stemness is maintained upon concurrent expression of RB and the mitochondrial ribosomal protein S18-2. Proc Natl Acad Sci U S A. 2020;117(27):1567315683. doi:10.1073/pnas.1922535117

45. Mallon BS, Hamilton RS, Kozhich OA, et al. Comparison of the molecular profiles of human embryonic and induced pluripotent stem cells of isogenic origin. Stem Cell Res. 2014;12(2):376386. doi:10.1016/j.scr.2013.11.010

46. de Almeida PE, Meyer EH, Kooreman NG, et al. Transplanted terminally differentiated induced pluripotent stem cells are accepted by immune mechanisms similar to self-tolerance. Nat Commun. 2014;5:3903. doi:10.1038/ncomms4903

47. Kooreman NG, Kim Y, de Almeida PE, et al. Autologous iPSC-based vaccines elicit anti-tumor responses in vivo. Cell Stem Cell. 2018;22(4):50113 e7. doi:10.1016/j.stem.2018.01.016

48. Medvedev SP, Shevchenko AI, Zakian SM. Induced pluripotent stem cells: problems and advantages when applying them in regenerative medicine. Acta Naturae. 2010;2(2):1828. doi:10.32607/20758251-2010-2-2-18-27

49. Lin M, Chang AE, Wicha M, Li Q, Huang S. Development and application of cancer stem cell-targeted vaccine in cancer immunotherapy. J Vaccines Vaccin. 2017;8(6). doi:10.4172/2157-7560.1000371

50. Ning N, Pan Q, Zheng F, et al. Cancer stem cell vaccination confers significant antitumor immunity. Cancer Res. 2012;72(7):18531864. doi:10.1158/0008-5472.CAN-11-1400

51. Chu DT, Nguyen TT, Tien NLB, et al. Recent progress of stem cell therapy in cancer treatment: molecular mechanisms and potential applications. Cells. 2020;9(3):563. doi:10.3390/cells9030563

52. Gabka-Buszek A, Kwiatkowska-Borowczyk E, Jankowski J, Kozlowska AK, Mackiewicz A. Novel genetic melanoma vaccines based on induced pluripotent stem cells or melanosphere-derived stem-like cells display high efficacy in a murine tumor rejection model. Vaccines (Basel). 2020;8(2). doi:10.3390/vaccines8020147

53. Wang P, Aguirre A. New strategies and in vivo monitoring methods for stem cell-based anticancer therapies. Stem Cells Int. 2018;2018:7315218. doi:10.1155/2018/7315218

54. Vanden Berg-Foels WS. In situ tissue regeneration: chemoattractants for endogenous stem cell recruitment. Tissue Eng Part B Rev. 2014;20(1):2839. doi:10.1089/ten.teb.2013.0100

55. Wang P, Moore A. Molecular imaging of stem cell transplantation for neurodegenerative diseases. Curr Pharm Des. 2012;18(28):44264440. doi:10.2174/138161212802481255

56. Hocking AM. The role of chemokines in mesenchymal stem cell homing to wounds. Adv Wound Care (New Rochelle). 2015;4(11):623630. doi:10.1089/wound.2014.0579

57. Ren G, Roberts AI, Shi Y. Adhesion molecules: key players in Mesenchymal stem cell-mediated immunosuppression. Cell Adh Migr. 2011;5(1):2022. doi:10.4161/cam.5.1.13491

58. Curfs JH, Meis JF, Hoogkamp-Korstanje JA. A primer on cytokines: sources, receptors, effects, and inducers. Clin Microbiol Rev. 1997;10(4):742780. doi:10.1128/CMR.10.4.742

59. Nakamizo A, Marini F, Amano T, et al. Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 2005;65(8):33073318. doi:10.1158/0008-5472.CAN-04-1874

60. Malekshah OM, Chen X, Nomani A, Sarkar S, Hatefi A. Enzyme/prodrug systems for cancer gene therapy. Curr Pharmacol Rep. 2016;2(6):299308. doi:10.1007/s40495-016-0073-y

61. Hasan A, Deeb G, Rahal R, et al. Mesenchymal stem cells in the treatment of traumatic brain injury. Front Neurol. 2017;8:28. doi:10.3389/fneur.2017.00028

62. Rosenblum D, Joshi N, Tao W, Karp JM, Peer D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat Commun. 2018;9(1):1410. doi:10.1038/s41467-018-03705-y

63. Layek B, Sadhukha T, Panyam J, Prabha S. Nano-engineered mesenchymal stem cells increase therapeutic efficacy of anticancer drug through true active tumor targeting. Mol Cancer Ther. 2018;17(6):11961206. doi:10.1158/1535-7163.MCT-17-0682

64. Bexell D, Gunnarsson S, Svensson A, et al. Rat multipotent mesenchymal stromal cells lack long-distance tropism to 3 different rat glioma models. Neurosurgery. 2012;70(3):731739. doi:10.1227/NEU.0b013e318232dedd

65. Cheng H, Kastrup CJ, Ramanathan R, et al. Nanoparticulate cellular patches for cell-mediated tumoritropic delivery. ACS Nano. 2010;4(2):625631. doi:10.1021/nn901319y

66. Chambers E, Mitragotri S. Long circulating nanoparticles via adhesion on red blood cells: mechanism and extended circulation. Exp Biol Med (Maywood). 2007;232(7):958966.

67. Hoshyar N, Gray S, Han H, Bao G. The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction. Nanomedicine (Lond). 2016;11(6):673692. doi:10.2217/nnm.16.5

68. Wang P, Yoo B, Sherman S, et al. Predictive imaging of chemotherapeutic response in a transgenic mouse model of pancreatic cancer. Int J Cancer. 2016;139(3):712718. doi:10.1002/ijc.30098

69. Zhao H, Richardson R, Talebloo N, Mukherjee P, Wang P, Moore A. uMUC1-targeting magnetic resonance imaging of therapeutic response in an orthotropic mouse model of colon cancer. Mol Imaging Biol. 2019;21(5):852860. doi:10.1007/s11307-019-01326-5

70. Zhao H, Hayat H, Ma X, Fan D, Wang P, Moore A. Molecular imaging and deep learning analysis of uMUC1 expression in response to chemotherapy in an orthotopic model of ovarian cancer. Sci Rep. 2020;10(1):14942. doi:10.1038/s41598-020-71890-2

71. Nouri FS, Wang X, Hatefi A. Genetically engineered theranostic mesenchymal stem cells for the evaluation of the anticancer efficacy of enzyme/prodrug systems. J Control Release. 2015;200:179187. doi:10.1016/j.jconrel.2015.01.003

72. Liu D, Hong Y, Li Y, et al. Targeted destruction of cancer stem cells using multifunctional magnetic nanoparticles that enable combined hyperthermia and chemotherapy. Theranostics. 2020;10(3):11811196. doi:10.7150/thno.38989

73. Dewhirst MW, Vujaskovic Z, Jones E, Thrall D. Re-setting the biologic rationale for thermal therapy. Int J Hyperthermia. 2005;21(8):779790. doi:10.1080/02656730500271668

74. Calderwood SK, Theriault JR, Gong J. How is the immune response affected by hyperthermia and heat shock proteins? Int J Hyperthermia. 2005;21(8):713716. doi:10.1080/02656730500340794

75. Wells J, Twamley S, Sekar A, et al. Lissajous scanning magnetic particle imaging as a multifunctional platform for magnetic hyperthermia therapy. Nanoscale. 2020;12(35):1834218355. doi:10.1039/D0NR00604A

76. Khizar S, Ahmad NM, Ahmed N, et al. Aminodextran coated CoFe2O4 nanoparticles for combined magnetic resonance imaging and hyperthermia. Nanomaterials (Basel). 2020;10(11):2182. doi:10.3390/nano10112182

77. Brennan G, Bergamino S, Pescio M, Tofail SAM, Silien C. The effects of a varied gold shell thickness on iron oxide nanoparticle cores in magnetic manipulation, T1 and T2 MRI contrasting, and magnetic hyperthermia. Nanomaterials (Basel). 2020;10(12):2424. doi:10.3390/nano10122424

See the article here:
The role of stem cells in cancer progression and therapy | OTT - Dove Medical Press

Acupuncture, Herbal Therapies Find Acceptance in Veterinary Care – The Two River Times

By Gloria Stravelli

LITTLE SILVER For Richard Yacowitz, D.V.M., of Little Silver Animal Hospital, adding integrative therapies like acupuncture, therapeutic laser and Chinese herbs to his practice is in line with a multidisciplinary approach to veterinary medicine.

Just because theres never one answer to a problem, he said of being open to integrative treatment options. I think that if you just keep an open mind in medicine, you can see things that you can add to your practice repertoire.

I started to offer acupuncture as I learned more and more about it, he said. Yacowitz received certification from the International Veterinary Acupuncture Society in 1996.

Once I was done with that, I started to practice, he said, adding Little Silver Animal Hospital was one of the first local veterinary practices to offer acupuncture.

And some pet owners were open to the integrative focus, he said.

It was relatively new but there were people open to the idea of trying it, he said, recalling a particular case involving a skeptical owner.

I remember there was a well-known judge in the area who lived in Rumson, Yacowitz recounted.

He brought me his dog and I said why dont we try acupuncture? I had just finished my course and he was known to be a very tough guy and he said, OK, well try it, but you know, Im a judge and I can make your life miserable.

So I said, Nah, youre not going to make my life miserable. Give it a try. If it works, it works and it worked.

In line with his integrative focus, Yacowitz employs other complementary therapies.

I always use a multimodal approach, other things that dont rely on just acupuncture or just cosequin (a nutritional supplement for animals). Theres also therapeutic laser and Chinese herbal medicine. I never rely on one thing.

According to Yacowitz, many pet owners are open to the integrative therapies, which in addition to acupuncture and herbs, include stem cell therapy to address osteoarthritis, joint damage and chronic allergies and diseases.

Veterinary medicine is always advancing, Yacowitz said. Were way ahead of the human medicine, because we can do things. We dont have to have 10-year studies with 100,000 patients. We can simply try things.

Research is great, but we dont have that ability to get the very big numbers you need but we do have lots and lots of veterinary research going on all over the world.

Another local practice embraced integrative therapies when holistic veterinarian Alexia Tsakiris, who holds a Bachelor of Veterinary Medicine, took a class in acupuncture that set her practice on a new path.

I saw how much it was helping me and went to a conference and learning about it sparked my interest, she said.

In 2016, Tsakiris opened Blue Sage Veterinary Wellness Center in Little Silver, incorporating both traditional and integrative treatments for pet patients.

I was doing conventional medicine alongside using a couple of herbs and I realized the time had come, she said. We are blessed to live in an area where there are many conventional veterinarians who do fantastic jobs in what they do. I wanted to be able to solely focus on what my passions are, which are acupuncture and herbs and holistic modalities.

While some were skeptical, all of her patients stayed with the practice andsupport also came from the local veterinary community.

I was better able to build relationships with veterinarians in the area because they knew what we were doing and they could see how it was helping the patients and everybody is able to do their best medicine in the interests of the pet, Tsakiris said.

She has found pet owners are open to the new therapies, particularly in difficult cases.

I had this one little dog who couldnt move, she said, he was paralyzed from the neck down. I said, I cant pretend Im a miracle worker but Im going to do my best. It took two weeks, and he started to move his head, by three weeks he was able to stand up and now the little guy is running around and its amazing.

It is even amazing for me sometimes, she said about the results she achieves.

According to Tsakiris, acupuncture addresses a broad range of pet health issues from arthritis and back issues to chronic issues and complements traditional treatments.

Thats where they start to thrive in these chronic conditions where conventional medicine doesnt have any more answers, she said.

Thats where acupuncture and herbs can really start to help even more. The treatments can help for chronic kidney disease, skin issues, behavior and cancers. The thing is that what I do can integrate with whatever the conventional vet is doing or we can do it alone. I take that case by case.

In addition to acupuncture, Tsakiris recently earned certification as a registered herbalist.

My real big focus is on traditional Chinese herbal formulas. Its an interplay of all of the different herbs from all of the different traditions and now all of the scientific research behind them.

The benefit of adding herbs with acupuncture, she said, is being able to tailor treatment to patient needs.

So, Im looking at the whole body and not just at the skin or an ear infection, she said. Im looking at the underlying causes and Im better able to treat those root causes that are creating the disease processes.

The article originally appeared in the April 15 21, 2021 print edition of The Two River Times.

View original post here:
Acupuncture, Herbal Therapies Find Acceptance in Veterinary Care - The Two River Times

Desperate 300,000 bid to save Easington Lane dad-of-three fighting cancer – Chronicle Live

A dad-of-three has launched a fundraising bid to fight his cancer after being told he has just months to live.

Derek Allen, of Easington Lane, near Houghton-le-Spring, was diagnosed with multiple myeloma - a form of blood cancer - in February 2019 after experiencing chest pains.

The 41-year-old was told his cancer could be treated, but not cured and was left stunned by the news that he would die.

He has since gone through five separate chemotherapy treatments and a stem cell transplant in his battle. And he is determined to fight on and receive the specialist treatment he needs.

He was initially given a life expectancy of seven years but treatments haven't worked and he now only has months to live.

Now he and his loving family have launched a 300,000 fundraising bid for specialist treatment in America

He said: "When I was first told about the blood cancer it felt like I had the rug pulled from underneath me.

"I'm a hardworking family man and I've always worked all week to provide for my family.

"I was frightened and scared, and I was trying to hold it together but my wife was crying. I was trying my best to be brave for her, but once I came out the room my head was full of so many questions.

Derek admits he is still coming to terms with his life drastically being cut short after receiving the shocking news.

He said: "I was relishing the treatment and said 'I'm going to prove everybody wrong and get through it all', but I've failed at every chance so far."

The dad-of-three, who works in steel fabrication and owns a window cleaning business, said he visited the doctor in early 2019 after repeated chest pains first experienced the year before.

He said: "One day at my nephew's wedding I had a sharp electrical pain in my ribs and I wasn't sure what it was.

"I thought it was down to the manual heavy labour work I did, and I felt like I was getting better, but weeks later it came back.

"I knew then it was time to go to the doctors and get it checked out."

After an initial x-ray Derek was rushed back to hospital for an emergency CT scan, which identified the multiple myeloma.

And he admits he has been fighting a constant battle since then after repeated setbacks with his treatment.

He added: "I've recently received results of a PET scan and they show new areas of medullary disease."

His attempts to gain access to clinical trials have been unsuccessful due to legislation and the NHS are unable to fund anymore treatment for him.

Now he sees specialist treatment in the United States as his last chance saloon.

Grandad Derek, who has been supported by his wife Catherine and three children Morgan 22, James 20 and Grace, 15, has so far raised over 8,000 for CAR-T therapy in the United States.

The diagnosis has also seen him value his life and family time a lot more, especially with grandson, Kayden, three, knowing that one day the cancer will defeat him.

"The news was hard for the kids initially but they're starting to come to terms with it," he said.

Hundreds of family, friends and strangers have donated to the fundraiser in a bid to support Derek.

He added: "I've been through all this and I haven't shed a tear in the last two years.

"But I get too emotional when I look at the fundraiser page. It's amazing to see the generosity."

Read the rest here:
Desperate 300,000 bid to save Easington Lane dad-of-three fighting cancer - Chronicle Live

Lake in the Hills police officer and father of 4 kids battling rare cancer forced to retire – Lake and McHenry County Scanner

Lake in the Hills Police Officer Mike Domagala with one of his children. | Provided Photo.

A Lake in the Hills police officer with 20 years of service, who is a father of four kids, says he will no longer be able to work as he battles a rare blood cancer, and a fundraiser has been started for him.

Mike Domagala began his law enforcement career in Fox River Grove in 2002. He was hired by the Lake in the Hills Police Department in 2012.

Domagala, 43, was diagnosed in July with multiple myeloma, which is a rare blood cancer that is not curable but is treatable.

According to the Mayo Clinic, cancerous plasma cells accumulate in the bone marrow and crowd out healthy blood cells. The cancer cells produce abnormal proteins that can cause complications.

Domagala is married and has four children, ages 5, 12, 16, and 23. At the onset of the cancer discovery, he had to have a pelvic biopsy, two bone marrow biopsies, blood work, PET scans, CT scans, and MRIs, according to a GoFundMe account.

Domagala has undergone multiple cycles of chemotherapy that started on August 4 and then a stem cell transplant in December.

He has not been able to patrol as an officer since his diagnosis. On March 25, Domagala said in an update that he had his 100-day post bone marrow appointment, which went well.

I also had my hematologist appointment for my maintenance therapy which will be a chemo pill every day and a bone healing transfusion once a month. The overwhelming support my family and I have received through this has been amazing and helps me continue to fight through this difficult time, he said.

Domagala said in February that he was optimistic about getting back to work. However, he said in his March 25 update that his doctor told him he would not be able to return to working the streets as a police officer.

I have been a police officer for almost 20 years and have always wanted to be since I was a child. I have no idea what I am going to do but I will figure it out as this is still sinking in, he said.

Fellow police officer Erik Watters started a GoFundMe account for Domagala in November and it continues to bring in donations from the public.

The fundraiser money is going towards uncovered medical expenses, travel expenses for treatments and to help his family with everyday expenses.

Mike maintains a strong fighting spirit and finds his strength in his love for his family. Mikes greatest concern is in continuing to provide for his family while he covers all of his uncovered medical expenses, Watter said.

The GoFundMe has a $50,000 goal and has raised $28,800 so far as of Thursday.

Follow this link:
Lake in the Hills police officer and father of 4 kids battling rare cancer forced to retire - Lake and McHenry County Scanner

Brentuximab Vedotin Plus Chemotherapy Works as a Primary Option for Hodgkin Lymphoma – Targeted Oncology

Pierluigi Porcu, MD, director, Medical Oncology and Hematopoietic Stem Cell Transplantation and coleader, Immune Cell Regulation and Targeting Program, Sidney Kimmel Cancer Center, at Jefferson Health in Philadelphia, PA, explained how the combination of brentuximab vedotin plus chemotherapy works in in frontline Hodgkin lymphoma.

Targeted OncologyTM: Following a diagnosis of classical Hodgkin lymphoma, nodular sclerosis type, what additional molecular testing should be ordered?

PORCU: PD-1 and PD-L1; I think most hematopathology labs nowadays run PD-1 and PD-L1 on Hodgkin [lymphoma] cases. Certainly, most hematopathology labs run CD30, although they may not run it on non-Hodgkin lymphoma cases on a routine basis.

Barr virus [EBV]. Its an in situ hybridization test thats fairly routine and not very expensive. It identifies cases of Hodgkin lymphoma that are EBV positive. Its important because there are some pretty good data that [show] EBV identifies a subgroup of Hodgkin lymphoma that has an inferior prognosis in terms of progression-free survival [PFS]. [Its also important because] there are now treatments for relapsed patients who have EBV-positive lymphoma including Hodgkin.

In some cases, the diagnosis is complex. You may have a gray-zone lymphoma or primary mediastinal B-cell lymphoma, [so its important] to have additional markers more in the space of diffuse large B-cell lymphoma to make sure.

In very difficult cases, the other testing would be immunoglobulin heavy chain gene rearrangement. This is routinely negative in Hodgkin lymphoma because the immunoglobulin in genes is aberrantly rearranged and mutated.

Why is PD-1 testing needed for diagnosis?

It doesnt affect diagnosis. Its part of the characterization of the Hodgkin lymphoma as a whole. I use it as a routine initial assessment because you never know when patients come back to have a second biopsy how easy it [will be] to get the tissue the second time around.

Can you discuss the International Prognostic Score [IPS]?

The IPS, [also called the Hasenclever index], was published in 1998 and is specifically for advanced-stage serum albumin level of less than 4; a hemoglobin level of less than 10.5; male sex; age equal to or older than 45; stage IV disease; and total WBC count of more than 15,000 or a lymphocyte count of less than 600, less than 8% of the WBC count, or both.1 The patient here has a number of these items. She has a hemoglobin of 9.5, stage IV disease, a WBC count higher than 15,000, and a lymphocyte count less than 600. Her IPS is 4. Based on the data from Hasenclever, her 5-year overall survival [OS] rate is 61%. Interestingly enough, [British Columbia Cancer] looked at their data in terms of the Hasenclever classes and found that for the patients with the highest scores, over time, some of these 5-year survivals were better, but not for the lower ones.

In addition, even though the lymphocyte count is part of the canonical standard, lymphopenia is a common phenomenon in most lymphomas. I follow it closely in T-cell lymphomas, where its common for people to have lymphocyte counts less than 500 and certainly less than 250 in AIDS territory, even though they dont have a history of opportunistic infection. I think the lymphocyte count is clearly an important biological flag marker, although we dont quite know what the biology of that is.

What are your thoughts on the options provided in the poll and the results?

Essentially here, were talking about SWOG S0816 [NCT00822120] or the RATHL [NCT00678327] clinical trial [regimens]. Straight-up escalated BEACOPP [bleomycin, etoposide, doxorubicin (Adriamycin), cyclophosphamide, vincristine (Oncovin), procarbazine, prednisone] German style, brentuximab vedotin [BV; Adcetris] with AVD [doxorubicin, vinblastine, dacarbazine] according to the ECHELON-1 [NCT01712490] trial, or other.

There were 4 votes for PET-adapted therapy with ABVD [doxorubicin, bleomycin, vinblastine, dacarbazine]. We have 7 who voted for BV plus AVD. No votes for escalated BEACOPP, which, even though its perfectly appropriate, Im happy to see [no votes for] because Im not a fan of escalated BEACOPP, and no [votes for] other. The majority would vote for the ECHELON-1 approach for this particular patient.

What frontline systemic therapy are you most likely to recommend for this patient?

Theres no role for radiation therapy. I think the jury is still out regarding PET-adapted therapy versus nonPET-adapted therapy. When I approach patients with advanced-stage disease, I look at a couple of things. One is, which risk category do they really belong to? The other is that PET-adapted therapy is dependent on the PET [scan]. Good-quality reading of PET scans is far from common; they dont give you Deauville [score]. Its unclear how they reach their conclusion because they dont compare the mediastinal and liver to the hypermetabolic lymph nodes. Its not that easy to get a good highquality PET interpretation, and if youre making these big decisions about the escalationor rather, in my case, deescalation according to the RATHL trialthen not having a goodquality PET is a big problem.

One of the big things about frontline therapy is that now we try to avoid exposure to bleomycin. There are 2 ways of doing that: Pick BV and AVD or treat the patient according to RATHL. Thats if the PET is negative; then you can deescalate and remove the bleomycin from the last 4 cycles of ABVD.2

Which factors do you think are most important?

It looks like everyone is on the same page as far as lung function and lung issues [if] someone is a heavy smoker or has a previous history of lung disease. Its not common in young people but certainly more common in middleaged or older people. Then, obviously, the selection of the therapy is important. The BV plus AVD had less lung toxicity compared to AVD, but its not that they had no lung toxicity. You still have to be worried somewhat and monitor people carefully on this therapy, even if it doesnt have bleomycin. Then for the PET-adapted ABVD, like RATHL, the first 2 cycles still contain bleomycin. Certainly, that is fundamental.

Here we have lung disease, prognostic score, performance. Age is really, in my practice, an important part of the decision-making because its not very common. Patients who are older than 60 or 65 generally represent no more than 20% of the cases of Hodgkin lymphoma. But when you have those patientsIm sure you all have seen them in your practicetheir prognosis is particularly bad, especially for those who are older than 70. Selecting the proper therapy for those patients is difficult and, of course, there are trials currently going on. Some of them have been published. For example, BV and bendamustine is one option. There are also trials with single-agent BV ongoing for patients who are frail on the front line. Besides, of course, all the combinations with checkpoint inhibitors...Theres quite a bit going on. For me, age is a very important component of decision-making.

Can you discuss the findings of the ECHELON-1 trial?

Following the initial phase 1 trial, data from ECHELON-1 showed that you cant give BV with ABVD because of a high rate of pulmonary toxicity. Finding the right dose of BV [is important] because it has to be given every 2 weeks. There are several dose-escalation records in phase 1, but 1.2 mg/kg was found to be the right tolerable dose for this schedule.

This was a large study of 1334 patients who were randomized 1:1 to receive 6 cycles of ABVD or 6 cycles of BV plus AVD. There was an interim PET scan, mostly to assess the response but not to be acted on, except for Deauville 5. [Those participants] would be given the opportunity to receive alternative therapy, and this was not part of the modified PFS scoring. Then there was an end of therapy CT-PET scan. Standard follow-up inclusion criteria [included] classical Hodgkin lymphoma that was stage III and IV, up to an ECOG performance status of 2, more than 18 years old, measurable disease, and adequate organ function. The primary end point was modified PFS, and a key secondary end point was OS.3

The initial paper was published in the New England Journal of Medicine in early 2018 with 2-year PFS data.3 But follow-up data [presented during the 2020 American Society of Hematology Annual Meeting and Exposition] show a median follow-up of 55.6 months with PFS of 82% [95% CI, 78.7%-84.8%] for BV plus AVD versus 75.2% [95% CI, 71.5%-78.4%] for ABVD. There is an advantage in terms of PFS.4 OS was no different between the 2 cohorts.

Not all the subgroups had a clear advantage, but younger patients, less than 45, and patients with the highest IPS did. There was an odd distinction: The North American cases appeared to have a stronger benefit compared with the European cases. I dont think anyone has a good explanation for that at this point. In addition, male sex and good performance status seem to be falling on the side of the fence that has a greater gain from BV plus AVD.5

In terms of adverse events [AEs], peripheral neuropathy was more common in the BV plus AVD group compared with the ABVD group [67% vs 43%, respectively]. There were also some gastrointestinal AEs, [including diarrhea (27% vs 18%, respectively) and abdominal pain (21% vs 10%)]. Overall, the 2 cohorts were fairly comparable in terms of overall AEs, except for...a greater number of hospitalizations and infections in the BV plus AVD cohort, which then led to the amendment toward the end of enrollment.3

Key toxicities are pulmonary toxicity and infections and neutropenia. Pulmonary toxicity was seen in both the BV plus AVD and ABVD cohorts, but ABVD had a significantly greater rate of pulmonary toxicity. In terms of on-study death, there were 9 deaths on the BV plus AVD cohort, and of those, 7 were from neutropenia or neutropenic infection. On the other hand, there were 13 deaths on the ABVD cohort, and the majority of them were because of pulmonary toxicity.3

Was the peripheral neuropathy reversible?

Eighty-four percent in the BV plus AVD cohort and 86% in the AVBD cohort reported complete resolution of peripheral neuropathy at almost 5 years. The median to improvement was 30 weeks for BV plus AVD and 16 weeks for AVBD, and then there was a subset that had ongoing neuropathy, mostly grades 1 and grade 2.4

In terms of survivorship, what guidance do you offer patients?

We still dont know much about safety from the standpoint of fertility. In this study, there were a good number of pregnancies and deliveries with healthy babies in women who participated.

Historically, there is a large body of data showing that the impact of ABVD, particularly 6 cycles of ABVD, is trivial to minimal in terms of fertility. Therefore, for patients treated with ABVD, I only [recommend] fertility consultation if the patient or the spouse has a significant degree of anxiousness about it. The other thing I always tell patients about fertilityand this is true for any diseaseis that its impossible to figure out what your fertility is if you never had kids. If someone had children already, then you know that their baseline fertility is standard or average. Its there. If they never did, then its impossible to say what it will be following treatment with anything.

We also dont know what the impact is going to be on second-line efficacy. BV is not going to be a weapon in your toolbox when people progress after BV plus AVD. I think that these are the big questions that we have that require long-term followup. Im selective with the patients that I treat with BV plus AVD. Certainly, for the younger patients, less than age 45, and patients who have advanced stage or high IPS scores, I tend to use this frontline approach. For the others, Im less enthusiastic about using it. I think the financial cost is worth it in those subgroups, including the risk to some degree, but it may not be in the others.

Is progressive multifocal leukoencephalopathy [PML] a concern?

Yes, there is some concern. Im very familiar with the PML cases that were diagnosed in patients who were treated with BV as a single agent with T-cell lymphoma. This is something that I mention to patients as a possible long-term, very severe risk, just like you have the same [risk] when you give Rituxan [rituximab]. I think there should be a concern, but its a minimal concern for this population based on the data that we have.

REFERENCES:

1. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkins disease. International Prognostic Factors Project on advanced Hodgkins disease. N Engl J Med. 1998;339(21):1506-1514. doi:10.1056/NEJM199811193392104

2. NCCN. Clinical Practice Guidelines in Oncology. Hodgkin lymphoma, version 2.2021. Accessed February 1, 2021. https://bit.ly/3oDAEZ4

3. Connors JM, Jurczak W, Straus DJ, et al; ECHELON-1 Study Group. Brentuximab vedotin with chemotherapy for stage III or IV Hodgkins lymphoma. N Engl J Med. 2018;378(4):331-344. doi:10.1056/NEJMoa1708984

4. Straus DJ, Dugosz-Danecka M, Connors J, et al. Brentuximab vedotin with chemotherapy for patients with previously untreated, stage III/IV classical Hodgkin lymphoma: 5-year update of the ECHELON-1 study. Presented at: 62nd American Society of Hematology Annual Meeting and Exposition; December 5-8, 2020; virtual. Accessed February 4, 2021. https://bit.ly/3pKKnx7

5. Straus DJ, Dugosz-Danecka M, Alekseev S, et al. Brentuximab vedotin with chemotherapy for stage III/IV classical Hodgkin lymphoma: 3-year update of the ECHELON-1 study. Blood. 2020;135(10):735-742. doi:10.1182/ blood.2019003127

Visit link:
Brentuximab Vedotin Plus Chemotherapy Works as a Primary Option for Hodgkin Lymphoma - Targeted Oncology

What are the Benefits of Regenerative Stem Cell Therapy …

Pet stem cell therapy is a process that has been used successfully on cats, dogs, and even horses. It is a branch of regenerative medicine that involves the treatment of bone and ligament injuries and osteoarthritis. And it can significantly improve your pets quality of life. Heres what you need to know about regenerative stem cell therapy in Texas.

Stem cells refer to undifferentiated cells whose daughter cells may easily differentiate into other cell types. This includes nerve, muscle, cartilage, fat, bone, liver, etc. They are used extensively in regenerative stem cell therapy.

Stem cells occur naturally in the body and are generally drawn to damaged areas. They swing into action by swiftly repairing the damaged tissue. In-depth studies show that stem cells contribute immensely to the repair of damaged bone tissue and cartilage. And they are known to assist in decreasing inflammation, preventing additional cell deterioration, minimizing pain, and promoting active tissue regeneration.

Pet stem cell therapy begins with the collection of cells or mesenchymal stromal from the bone marrow in the animals femur. They can also be taken from blood plasma.

These cells are then isolated and concentrated via a specializing system. as soon as this process is complete, the stem cells can be injected into affected joints or damaged tissue to promote the healing process.

There are presently no comprehensive studies that fully support the fact that pet stem cell therapy works. Several stories from pet owners or veterinarians prove that it works. Research is still ongoing to determine the efficacy of this procedure on pets.

But one profound benefit of regenerative stem cell therapy is that there are little or zero risks of rejection or reaction. This is because the stem cells are harvested from your pet. This means that the inflamed or damaged area will heal up faster as it quickly develops the characteristics of cells required to repair a pets injuries.

Stem cells can also jumpstart the healing process by activating nearby cells and even recruiting additional ones for tissue repair.

Any injured petcats, dogs, horses, etc.can benefit immensely from regenerative stem cell therapy. Therefore, if your canine or feline has arthritic joints or damaged tissue, pet stem cell therapy may be a viable option.

Other benefits of stem cell therapy include:

Clinical trials have shown that this medical approach can be adopted for injuries linked to the spinal cord. However, there is little concrete evidence that shows that stem cell therapy works as expected.

If your dog or pet has damaged tissue or joint or suffers from osteoarthritis, undergoing regenerative stem cell therapy is an ideal option.

Go here to learn more about stem cell therapy and taking your pet to a reliable stem cell therapy clinic in Texas.

Link:
What are the Benefits of Regenerative Stem Cell Therapy ...

China Mesenchymal Stem Cells Market Share, Growth Factors, Top Manufacturers, By Types, Applications And Forecast To 2027 Express Keeper – Express…

The global China Mesenchymal Stem Cells Market report by BMRC provides a detailed analysis of the area marketplace expanding; competitive landscape; global, regional, and country-level market size; market growth analysis; market share; opportunities analysis; product launches; recent developments; sales analysis; segmentation growth; technological innovations; and value chain optimization. The report offers a comprehensive list of key players, their strategies they adopt to sustain in the market. All of this and more information is covered in 200 pages.

The Coronavirus (COVID-19) pandemic has affected every aspect of life worldwide. It has forced various industries to re-evaluate their strategies and adopt new ones to sustain during these trying times. The latest report includes the current COVID-19 impact on the market.

Top Key players Thermo Fisher, Bio-Techne, ATCC, MilliporeSigma, PromoCell GmbH, Genlantis, Celprogen, Cell Applications, Cyagen Biosciences, Axol Bioscience

Get PDF Sample Brochure:https://brandessenceresearch.biz/Request/Sample?ResearchPostId=98050&RequestType=Sample

The global China Mesenchymal Stem Cells market report is a comprehensive analysis of the current and future analysis, which is based on historic data. This provides the reader with quantified data, enabling them to take well informed business decisions. The report has been written using primary and secondary research. It includes predictive analysis, Porters 5 force analysis, SWOT analysis, and real-time analytics. Several graphs have been provided to support the data and for a clear understanding of various facts and figures.

The report has been divided into product types, application, end-users, and regions. These segments provide accurate calculations and forecasts for sales in terms of volume and value. This analysis can help customers increase their business and to take calculated decisions.

The Global China Mesenchymal Stem Cells Market Report has been Segments into:

On the basis of product, this report displays the production, revenue, price, market share and growth rate of each type, primarily split into Human MSC, Mouse MSC, Rat MSC, Other

On the basis of the end users/applications, this report focuses on the status and outlook for major applications/end users, consumption (sales), market share and growth rate for each application, including Research Institute, Hospital, Others

Global China Mesenchymal Stem Cells Market Size & Share, By Regions and Countries/Sub-regions,

Asia Pacific: China, Japan, India, and Rest of Asia Pacific

Europe: Germany, the UK, France, and Rest of Europe

North America: the US, Mexico, and Canada

Latin America: Brazil and Rest of Latin America

Middle East & Africa: GCC Countries and Rest of Middle East & Africa

Research Objectives

To analyze and forecast the Worldwide China Mesenchymal Stem Cells, in terms of value and volume.

Which segment has the potential to gain the highest market share?

To help decision-makers from a new offer perspective and benchmark existing marketing strategy.

Correlate cost structure historical data with key business segments.

Analyze marketing contribution and customer acquisition by up-selling and cross-selling.

Identifying Influencing factors keeping Worldwide China Mesenchymal Stem Cells Intense, factored with periodic analysis of CR4 & CR8 concentration ratio & HHI Index.

Important Features that are under offering & key highlights of the report:

1) Does the study cover COVID-19 Impact Analysis and its effect on Growth %?

Yes, the overall industry has seen quite a big impact due to slowdown and shutdown in the production line & supply chain. The study covers a separate qualitative chapter on COVID-19 Impact analysis. Additionally, it also provides before and after the scenario of COVID-19 on sales growth & market size estimation to better analyze the exact scenario of the industry.

2) How companies are selected or profiled in the report?

List of some players that are profiled in the report include:

Thermo Fisher, Bio-Techne, ATCC, MilliporeSigma, PromoCell GmbH, Genlantis, Celprogen, Cell Applications, Cyagen Biosciences, Axol Bioscience

Usually, we follow NAICS Industry standards and validate company profile with product mapping to filter relevant Industry players, furthermore the list is sorted to come up with a sample size of at least 50 to 100 companies having greater topline value to get their segment revenue for market estimation.

** List of companies mentioned may vary in the final report subject to Name Change / Merger etc.

3) Can we add or profiled a new company as per our needs?

Yes, we can add or profile a new company as per client need in the report, provided it is available in our coverage list as mentioned in answer to Question 1 and after feasibility run, final confirmation will be provided by the research team checking the constraints related to the difficulty of survey.

4) Can we narrow the available business segments?

Yes, depending upon the data availability and feasibility check by our Research Analyst, a further breakdown in business segments by end-use application or product type can be provided (If applicable) by Revenue Size or Volume*.

China Mesenchymal Stem Cells market segmentation

Type Human MSC, Mouse MSC, Rat MSC, Other

Application Research Institute, Hospital, Others

5) Can a specific country of interest be added? What all regional segmentation covered?

Yes, Country-level splits can be modified in the study as per objectives. Currently, the research report gives special attention and focus on the following regions:

North America [United States, Canada, Mexico], Asia-Pacific [China, India, Japan, South Korea, Australia, Indonesia, Malaysia, Philippines, Thailand, Vietnam], Europe [Germany, France, UK, Italy, Russia, Rest of Europe], South America [Brazil, Argentina, Rest of South America], Middle East & Africa [GCC Countries, Turkey, Egypt, South Africa, Rest of the Middle East & Africa]

** One country of specific interest can be included at no added cost. For inclusion of more regional segment quotes will vary.

Request Customization of the Report: https://brandessenceresearch.biz/Request/Sample?ResearchPostId=98050&RequestType=Methodology

Table of Contents1 Industry Overview of Pet Insurance1.1 Brief Introduction of Pet Insurance1.1.1 Definition of Pet Insurance1.1.2 Development of China Mesenchymal Stem CellsIndustry1.2 Classification of Pet Insurance1.3 Status of China Mesenchymal Stem CellsIndustry1.3.1 Industry Overview of Pet Insurance1.3.2 Global Major Regions Status of Pet Insurance

2 Industry Chain Analysis of Pet Insurance2.1 Supply Chain Relationship Analysis of Pet Insurance2.2 Upstream Major Raw Materials and Price Analysis of Pet Insurance2.3 Downstream Applications of Pet Insurance

3 Manufacturing Technology of Pet Insurance3.1 Development of China Mesenchymal Stem CellsManufacturing Technology3.2 Manufacturing Process Analysis of Pet Insurance3.3 Trends of China Mesenchymal Stem CellsManufacturing Technology

4 Major Manufacturers Analysis of Pet Insurance4.1 Company 14.1.1 Company Profile4.1.2 Product Picture and Specifications4.1.3 Capacity, Production, Price, Cost, Gross and Revenue4.1.4 Contact Information4.2 Company 24.2.1 Company Profile4.2.2 Product Picture and Specifications4.2.3 Capacity, Production, Price, Cost, Gross and Revenue4.2.4 Contact Information4.3 Company 34.3.1 Company Profile4.3.2 Product Picture and Specifications4.3.3 Capacity, Production, Price, Cost, Gross and Revenue4.3.4 Contact Information4.4 Company 44.4.1 Company Profile4.4.2 Product Picture and Specifications4.4.3 Capacity, Production, Price, Cost, Gross and Revenue4.4.4 Contact Information4.5 Company 54.5.1 Company Profile4.5.2 Product Picture and Specifications4.5.3 Capacity, Production, Price, Cost, Gross and Revenue4.5.4 Contact Information4.6 Company 64.6.1 Company Profile4.6.2 Product Picture and Specifications4.6.3 Capacity, Production, Price, Cost, Gross and Revenue4.6.4 Contact Information4.7 Company 74.7.1 Company Profile4.7.2 Product Picture and Specifications4.7.3 Capacity, Production, Price, Cost, Gross and Revenue4.7.4 Contact Information4.8 Company 84.8.1 Company Profile4.8.2 Product Picture and Specifications4.8.3 Capacity, Production, Price, Cost, Gross and Revenue4.8.4 Contact Information4.9 Company 94.9.1 Company Profile4.9.2 Product Picture and Specifications4.9.3 Capacity, Production, Price, Cost, Gross and Revenue4.9.4 Contact Information4.10 Company ten4.10.1 Company Profile4.10.2 Product Picture and Specifications4.10.3 Capacity, Production, Price, Cost, Gross and Revenue4.10.4 Contact Information. . .

5 Global Productions, Revenue and Price Analysis of China Mesenchymal Stem Cellsby Regions, Manufacturers, Types and Applications5.1 Global Production, Revenue of China Mesenchymal Stem Cellsby Regions 2014-20195.2 Global Production, Revenue of China Mesenchymal Stem Cellsby Manufacturers 2014-20195.3 Global Production, Revenue of China Mesenchymal Stem Cellsby Types 2014-20195.4 Global Production, Revenue of China Mesenchymal Stem Cellsby Applications 2014-20195.5 Price Analysis of Global China Mesenchymal Stem Cellsby Regions, Manufacturers, Types and Applications in 2014-2019

6 Global and Major Regions Capacity, Production, Revenue and Growth Rate of China Mesenchymal Stem Cells2014-20196.1 Global Capacity, Production, Price, Cost, Revenue, of China Mesenchymal Stem Cells2014-20196.2 Asia Pacific Capacity, Production, Price, Cost, Revenue, of China Mesenchymal Stem Cells2014-20196.3 Europe Capacity, Production, Price, Cost, Revenue, of China Mesenchymal Stem Cells2014-20196.4 Middle East & Africa Capacity, Production, Price, Cost, Revenue, of China Mesenchymal Stem Cells2014-20196.5 North America Capacity, Production, Price, Cost, Revenue, of China Mesenchymal Stem Cells2014-20196.6 Latin America Capacity, Production, Price, Cost, Revenue, of China Mesenchymal Stem Cells2014-2019

7 Consumption Volumes, Consumption Value, Import, Export and Sale Price Analysis of China Mesenchymal Stem Cellsby Regions7.1 Global Consumption Volume and Consumption Value of China Mesenchymal Stem Cellsby Regions 2014-20197.2 Global Consumption Volume, Consumption Value and Growth Rate of China Mesenchymal Stem Cells2014-20197.3 Asia Pacific Consumption Volume, Consumption Value, Import, Export and Growth Rate of China Mesenchymal Stem Cells2014-20197.4 Europe Consumption Volume, Consumption Value, Import, Export and Growth Rate of China Mesenchymal Stem Cells2014-20197.5 Middle East & Africa Consumption Volume, Consumption Value, Import, Export and Growth Rate of China Mesenchymal Stem Cells2014-20197.6 North America Consumption Volume, Consumption Value, Import, Export and Growth Rate of China Mesenchymal Stem Cells2014-20197.7 Latin America Consumption Volume, Consumption Value, Import, Export and Growth Rate of China Mesenchymal Stem Cells2014-20197.8 Sale Price Analysis of Global China Mesenchymal Stem Cellsby Regions 2014-2019

8 Gross and Gross Margin Analysis of Pet Insurance8.1 Global Gross and Gross Margin of China Mesenchymal Stem Cellsby Regions 2014-20198.2 Global Gross and Gross Margin of China Mesenchymal Stem Cellsby Manufacturers 2014-20198.3 Global Gross and Gross Margin of China Mesenchymal Stem Cellsby Types 2014-20198.4 Global Gross and Gross Margin of China Mesenchymal Stem Cellsby Applications 2014-2019

9 Marketing Traders or Distributor Analysis of Pet Insurance9.1 Marketing Channels Status of Pet Insurance9.2 Marketing Channels Characteristic of Pet Insurance9.3 Marketing Channels Development Trend of Pet Insurance

10 Global and Chinese Economic Impacts on China Mesenchymal Stem CellsIndustry10.1 Global and Chinese Macroeconomic Environment Analysis10.1.1 Global Macroeconomic Analysis and Outlook10.1.2 Chinese Macroeconomic Analysis and Outlook10.2 Effects to China Mesenchymal Stem CellsIndustry

11 Development Trend Analysis of Pet Insurance11.1 Capacity, Production and Revenue Forecast of China Mesenchymal Stem Cellsby Regions, Types and Applications11.1.1 Global Capacity, Production and Revenue of China Mesenchymal Stem Cellsby Regions 2019-202411.1.2 Global and Major Regions Capacity, Production, Revenue and Growth Rate of China Mesenchymal Stem Cells2019-202411.1.3 Global Capacity, Production and Revenue of China Mesenchymal Stem Cellsby Types 2019-202411.2 Consumption Volume and Consumption Value Forecast of China Mesenchymal Stem Cellsby Regions11.2.1 Global Consumption Volume and Consumption Value of China Mesenchymal Stem Cellsby Regions 2019-202411.2.2 Global and Major Regions Consumption Volume, Consumption Value and Growth Rate of China Mesenchymal Stem Cells2019-202411.3 Supply, Import, Export and Consumption Forecast of Pet Insurance11.3.1 Supply, Consumption and Gap of China Mesenchymal Stem Cells2019-202411.3.2 Global Capacity, Production, Price, Cost, Revenue, Supply, Import, Export and Consumption of China Mesenchymal Stem Cells2019-202411.3.3 North America Capacity, Production, Price, Cost, Revenue, Supply, Import, Export and Consumption of China Mesenchymal Stem Cells2019-202411.3.4 Europe Capacity, Production, Price, Cost, Revenue, Supply, Import, Export and Consumption of China Mesenchymal Stem Cells2019-202411.3.5 Asia Pacific Capacity, Production, Price, Cost, Revenue, Supply, Import, Export and Consumption of China Mesenchymal Stem Cells2019-202411.3.6 Middle East & Africa Capacity, Production, Price, Cost, Revenue, Supply, Import, Export and Consumption of China Mesenchymal Stem Cells2019-202411.3.7 Latin America Capacity, Production, Price, Cost, Revenue, Supply, Import, Export and Consumption of China Mesenchymal Stem Cells2019-2024

12 Contact information of Pet Insurance12.1 Upstream Major Raw Materials and Equipment Suppliers Analysis of Pet Insurance12.1.1 Major Raw Materials Suppliers with Contact Information Analysis of Pet Insurance12.1.2 Major Equipment Suppliers with Contact Information Analysis of Pet Insurance12.2 Downstream Major Consumers Analysis of Pet Insurance12.3 Major Suppliers of China Mesenchymal Stem Cellswith Contact Information12.4 Supply Chain Relationship Analysis of Pet Insurance

13 New Project Investment Feasibility Analysis of Pet Insurance13.1 New Project SWOT Analysis of Pet Insurance13.2 New Project Investment Feasibility Analysis of Pet Insurance13.2.1 Project Name13.2.2 Investment Budget13.2.3 Project Product Solutions13.2.4 Project Schedule

14 Conclusion of the Global China Mesenchymal Stem CellsIndustry 2019 Market Research Report

Listoftables

to be continued

Top Trending Reports:

https://www.marketwatch.com/press-release/europe-healthcare-edi-market-size-share-2021-latest-developments-growth-prospects-business-share-competitive-status-future-trends-by-regions-top-manufacturers-and-key-statistics-forecast-to-2027-2021-02-15?tesla=y

https://www.marketwatch.com/press-release/trends-in-baby-care-products-market-industry-analysis-and-opportunity-assessment-2021-2027-2021-02-17?tesla=y

https://www.marketwatch.com/press-release/phycocyanin-market-size-research-report-insights-and-forecast-2021-to-2027-2021-02-15?tesla=y

https://www.marketwatch.com/press-release/us-oligonucleotide-synthesis-market-trends-growth-opportunities-key-players-revenue-emerging-trends-and-business-strategy-till-2027-2021-02-15?tesla=y

https://www.marketwatch.com/press-release/lignin-market-global-outlook-2021-technology-trends-industry-growth-opportunities-size-estimation-top-company-share-regional-forecast-to-2027-2021-02-15?tesla=y

Here is the original post:
China Mesenchymal Stem Cells Market Share, Growth Factors, Top Manufacturers, By Types, Applications And Forecast To 2027 Express Keeper - Express...

In B-Cell NHL, Off-the-Shelf NK Immunotherapy Shows Promise When Used With Chemotherapy and ASCT – Cancer Network

Early results from a phase 2 trial (NCT03019640) showed that cord bloodderived natural killer (NK) immunotherapy plus high-dose chemotherapy and autologous stem cell transplant (ASCT) induced early antitumor response in B-cell non-Hodgkin lymphoma (NHL).

The trial investigators evaluated the potential of the novel cellular cord bloodderived NK immunotherapy in patients with B-cell NHL who are undergoing high-dose chemotherapy and ASCT and presented their findings in a poster at the recent 2021 Transplant & Cellular Therapy Meetings. They chose CB units for NK cell expansion on artificial antigen presenting cells and without human leukocyte antigens (HLA) matching to provide increased expansion and make the treatment off-the-shelf capable.

The study enrolled patients who were aged 15 to 70 with B-cell NHL, excluding primary central nervous system lymphoma, who were candidates for high-dose chemotherapy and ASCT. Patients were required to have adequate end-organ function, an ECOG performance status of 0 or 1, and prior apheresis of 2 x 106 CD34+ cells/Kg in order to be eligible.

Those with prior whole brain irradiation, active hepatitis B, evidence of cirrhosis or high-grade liver fibrosis, active infection, HIV infection, or received radiation therapy within the past month were excluded from joining the trial.

Patients were given intravenous (IV) carmustine over 2 hours 12 days prior to transplant, IV etoposide twice daily over 3 hours and IV cytarabine twice daily over 1 hour for days 11 to 8 prior to transplant, IV melphalan over 30 minutes 7 days prior to transplant, oral lenalidomide (Revlimid) once daily for days 7 through 2 prior to transplant. Additionally, patients who are CD20-positive received IV rituximab (Rituxan) over 3 hours for days 13 through 7 prior to transplant.

Then patients received cord bloodderived expanded allogeneic NK cells intravenously over 1 hour on day 5 prior to ASCT. Five days following ASCT, patients started received daily subcutaneous filgrastim.

The primary end point was 30-day treatment-related mortality (TRM) and secondary end points were relapse-free survival (RFS), overall survival, and NK cell persistence.

Study authors, led by Yago L. Nieto, MD, PhD, in the Department of Stem Cell Transplantation, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center in Houston, provided an update on data for 20 of the enrolled patients in the study. Patients were enrolled between December 2017 and July 2020. One patient experienced rapid tumor progression during culture of NK cells and thus was not treated in the study.

The median age of the 19 treated patients was 60 years (range, 33-70) with the majority (73.7%) being male and having diffuse large B-cell lymphoma (DLBCL; 84.2%); the 3 remaining patients had mantle cell lymphoma (n = 2) or follicular lymphoma (n = 1). More than two-thirds (68.4%) of patients had relapsed disease whereas the 2 patients with MCL were being treated in the frontline setting and 4 patients had primary refractory disease. The median number of prior lines of therapy was 2 (range, 1-4).

Response assessed through PET at ASCT was a complete response for 78.9%, partial response for 15.8%, and progressive disease for 5.3%. Nine patients had 1/6 HLA match of cord blood at DR, 6 had 1/6 match at B, 3 had 1/6 match at A, and 1 had 2/6 match at B and DR.

Cord bloodderived NK cells were expanded a median of 1552-fold (range, 317-4767) with the infused NK product comprising a CD3-CD16+CD56+ phenotype for a median of 98.9% (range, 97.6%-99.5%) of the cells. The cord bloodderived NK cells had a median viability of 96.5% (range, 92%-98%). In the peripheral blood, NK cells were detectable for a mean of 2 weeks (range, 2-3).

For weeks 1 through 3, the cord bloodderived NK cells showed a higher percentage of NKG2D and NKp30 activation receptors than the patients own NK cells; the study authors noted that this indicated an effector phenotype. Additionally, NK persistence was found not to be impacted by the degree of HLA mismatch.

At a median follow-up of 18 months (range, 4-340), the RFS rate was 68% and the OS rate was 84%. Eleven of the 16 patients (68.8%) with DLBCL are still in remission.

The study authors concluded that expanded and highly purified cord bloodderived NK immunotherapy was safe and promising in combination with high-dose chemotherapy and ASCT in patients with B-cell NHL.

Reference:

Nato Y, Kaur I, Hosing C, et al. Immunotherapy with ex vivo-expanded cord blood (CB)-derived nk cells combined with high-dose chemotherapy (HDC) and autologous stem cell transplant (ASCT) for B-cell non-hodgkins lymphoma (NHL). Presented at: 2021 Transplant & Cellular Therapy Meetings; February 8-12, 2021; Virtual. LBA15.

See the original post here:
In B-Cell NHL, Off-the-Shelf NK Immunotherapy Shows Promise When Used With Chemotherapy and ASCT - Cancer Network

Back to Top