page contents

sites

Monday 4 February 2013

Signs Of Leukemia

Source(google.com.pk)
Signs Of Leukemia Biography

There are two types of leukemia, acute and chronic. In acute leukemia the disease progresses rapidly, there is an accumulation of immature, useless cells in the marrow and blood. In chronic leukemia, the disease develops more slowly and allows more mature, useful cells to be made. Acute leukemia crowds out the good cells more rapidly than chronic leukemia. "Acute" means "sudden onset", while "chronic" means "long-term".

Acute lymphoblastic leukemia, also known as acute lymphocytic leukemia is when the blood and bone marrow have large numbers of white blood cells destined to become lymphocytes.

Acute lymphoblastic leukemia is commonly referred to by the acronym ALL, pronounced as three separate letters - eigh, el, el. ALL is the most common childhood cancer, especially among toddlers aged 2 to 3 years.

In the USA, ALL incidence is significantly higher among Caucasian than African-American children. Incidence is highest among Hispanic children.

In the USA there are about 6,000 new cases of ALL annually, an incidence of 1 in every 50,000 people.

In England and Wales there are approximately 2,400 diagnosed cases of acute leukemia each year, of which about 600 are ALLs. The NHS (National Health Service), UK, informs that 85% of all ALL cases occur among children aged less than 15 years.

Experts say the main causes of ALL are exposure to high levels of radiation or benzene. Smokers are three times as likely to develop ALL compared to non-smokers - benzene is one of the 4,000 substances found in cigarette smoke. Individuals who have spent over 5,000 hours in airplanes have a higher risk of developing ALL (flying exposes you to more of the Sun's radiation). About 1 in every 20 cases are thought to be caused by related genetic disorders, such as Down's syndrome.

Some scientists wonder whether early exposure to germs might protect children from developing ALL. A significantly lower percentage of children who went to playgroups at an early age develop ALL compared to those who didn't. Symptoms of acute lymphoblastic leukemia will usually start slowly, and then escalate in severity as the number of blast cells in the blood rises. Signs and symptoms may include:
Fatigue
Frequent unexplained bleeding, such as nosebleeds or bleeding gums
High fever
Oversweating
Painful joints and/or bones
Panting
Several infections over a short period
Swollen glands (lymph nodes)
Swollen liver
Swollen spleen
The skin bruises easily
The skin is paler than it should be
Unexplained weight loss
If the affected cells spread into the CNS (central nervous system), the patient may have neurological symptoms, such as dizziness, vomiting, blurred vision, fits (seizures) and headaches.

With the right treatment virtually all children will become symptom free (remission) and 85% will be completely cured. Unfortunately, only about 40% of adult patients achieve a complete cure.

Treatment involves a combination of chemotherapy and radiotherapy (radiation therapy). Sometimes a bone marrow transplant may be used.

For those who are not cured, their immune systems, which are very low in white blood cells, become vulnerable to infections, some of them life-threatening. There is also a risk of serious bleeding due to a lack of platelets.

Approximately 230 people die from ALL in England and Wales each year.

Dutch athlete, Maarten van der Weijden, was diagnosed with ALL in 2001. He went on to win the 10 km open water marathon race at the 2008 Summer Olympics in Beijing.

Andrew McMahon, singer of the bands Something Corporate and Jack's Mannequin, was diagnosed with ALL in 2005.

Acute Leukemia

Source(google.com.pk)
Acute Leukemia Biography

Leukemia (British spelling: leukaemia ) is cancer of the blood or bone marrow (which produces blood cells). A person who has leukemia suffers from an abnormal production of blood cells, generally leukocytes (white blood cells).

The word Leukemia comes from the Greek leukos which means "white" and aima which means "blood".

The DNA of immature blood cells, mainly white cells, becomes damaged in some way. This abnormality causes the blood cells to grow and divide chaotically. Normal blood cells die after a while and are replaced by new cells which are produced in the bone marrow. The abnormal blood cells do not die so easily, and accumulate, occupying more and more space. As more and more space is occupied by these faulty blood cells there is less and less space for the normal cells - and the sufferer becomes ill. Quite simply, the bad cells crowd out the good cells in the blood.

In order to better understand what goes on we need to have a look at what the bone marrow does.

Function of the bone marrow
The bone marrow is found in the inside of bones. The marrow in the large bones of adults produces blood cells. Approximately 4% of our total bodyweight consists of bone marrow.

There are two types of bone marrow: 1. Red marrow, made up mainly of myeloid tissue. 2. Yellow marrow, made up mostly of fat cells. Red marrow can be found in the flat bones, such as the breast bone, skull, vertebrae, shoulder blades, hip bone and ribs. Red marrow can also be found at the ends of long bones, such as the humerus and femur.

White blood cells (lymphocytes), red blood cells and platelets are produced in the red marrow. Red blood cells carry oxygen, white blood cells fight diseases. Platelets are essential for blood clotting. Yellow marrow can be found in the inside of the middle section of long bones.

If a person loses a lot of blood the body can convert yellow marrow to red marrow in order to raise blood cell production.

White blood cells, red blood cells and platelets exist in plasma - Blood plasma is the liquid component of blood, in which the blood cells are suspended.
Types of leukemia

Chronic and Acute
Experts divide leukemia into four large groups, each of which can be Acute, which is a rapidly progressing disease that results in the accumulation of immature, useless cells in the marrow and blood, or Chronic, which progresses more slowly and allows more mature, useful cells to be made. In other words, acute leukemia crowds out the good cells more quickly than chronic leukemia.

Lymphocytic and Myelogenous
Leukemias are also subdivided into the type of affected blood cell. If the cancerous transformation occurs in the type of marrow that makes lymphocytes, the disease is called lymphocytic leukemia. A lymphocyte is a kind of white blood cell inside your vertebrae immune system. If the cancerous change occurs in the type of marrow cells that go on to produce red blood cells, other types of white cells, and platelets, the disease is called myelogenous leukemia.

Acute Lymphocytic Leukemia (ALL), also known as Acute Lymphoblastic Leukemia - This is the most common type of leukemia among young children, although adults can get it as well, especially those over the age of 65. Survival rates of at least five years range from 85% among children and 50% among adults. The following are all subtypes of this leukemia: precursor B acute lymphoblastic leukemia, precursor T acute lymphoblastic leukemia, Burkitt's leukemia, and acute biphenotypic leukemia.

Chronic Lymphocytic Leukemia (CLL) - This is most common among adults over 55, although younger adults can get it as well. CLL hardly ever affects children. The majority of patients with CLL are men, over 60%. 75% of treated CLL patients survive for over five years. Experts say CLL is incurable. A more aggressive form of CLL is B-cell prolymphocytic leukemia.

Acute Myelogenous Leukemia (AML) - AML is more common among adults than children, and affects males significantly more often than females. Patients are treated with chemotherapy. 40% of treated patients survive for over 5 years. The following are subtypes of AMS - acute promyelocytic leukemia, acute myeloblastic leukemia, and acute megakaryoblastic leukemia.

Researchers from the Memorial Sloan-Kettering Cancer Center reported in the March 2012 issue of NEJM (New England Journal of Medicine that they identified a series of genetic mutations in people with AML. They explained that their findings may help doctors to more accurately predict patient outcomes, as well as choosing therapies they are most likely to respond to.

Chronic Myelogenous Leukemia (CML) - The vast majority of patients are adults. 90% of treated patients survive for over 5 years. Gleevec (imatinib) is commonly used to treat CML, as well as some other drugs. Chronic monocytic leukemia is a subtype of CML.

Symptoms of leukemia
Blood clotting is poor - As immature white blood cells crowd out blood platelets, which are crucial for blood clotting, the patient may bruise or bleed easily and heal slowly - he may also develop petechiae (a small red to purple spot on the body, caused by a minor hemorrhage).

Affected immune system - The patient's white blood cells, which are crucial for fighting off infection, may be suppressed or not working properly. The patient may experience frequent infections, or his immune system may attack other good body cells.

Anemia - As the shortage of good red blood cells grows the patient may suffer from anemia - this may lead to difficult or labored respiration (dyspnea) and pallor (skin has a pale color caused by illness).

Other symptoms - Patients may also experience nausea, fever, chills, night sweats, flu-like symptoms, and tiredness. If the liver or spleen becomes enlarged the patient may feel full and will eat less, resulting in weight loss. Headache is more common among patients whose cancerous cells have invaded the CNS (central nervous system).

Precaution - As all these symptoms could be due to other illnesses. A diagnosis of leukemia can only be confirmed after medical tests are carried out.

What causes leukemia?
Experts say that different leukemias have different causes. The following are either known causes, or strongly suspected causes:
Artificial ionizing radiation
Viruses - HTLV-1 (human T-lymphotropic virus) and HIV (human immunodeficiency virus)
Benzene and some petrochemicals
Alkylating chemotherapy agents used in previous cancers
Maternal fetal transmission (rare)
Hair dyes
Genetic predisposition - some studies researching family history and looking at twins have indicated that some people have a higher risk of developing leukemia because of a single gene or multiple genes.
Down syndrome - people with Down syndrome have a significantly higher risk of developing leukemia, compared to people who do not have Down syndrome. Experts say that because of this, people with certain chromosomal abnormalities may have a higher risk.
Electromagnetic energy - studies indicate there is not enough evidence to show that ELF magnetic (not electric) fields that exist currently might cause leukemia. The IARC (International Agency for Research on Cancer) says that studies which indicate there is a risk tend to be biased and unreliable.

Treatments for leukemia
As the various types of leukemias affect patients differently, their treatments depend on what type of leukemia they have. The type of treatment will also depend on the patient's age and his state of health.

In order to get the most effective treatment the patient should get treatment at a center where doctors have experience and are well trained in treating leukemia patients. As treatment has improved, the aim of virtually all health care professionals should be complete remission - that the cancer goes away completely for a minimum of five years after treatment.

Treatment for patients with acute leukemias should start as soon as possible - this usually involves induction therapy with chemotherapy, and takes place in a hospital.

When a patient is in remission he will still need consolidation therapy or post induction therapy. This may involve chemotherapy, as well as a bone marrow transplant (allogeneic stem cell transplantation).

If a patient has Chronic Myelogenous Leukemia (CML) his treatment should start as soon as the diagnosis is confirmed. He will be given a drug, probably Gleevec (imatinib mesylate), which blocks the BCR-ABL cancer gene. Gleevec stops the CML from getting worse, but does not cure it. There are other drugs, such as Sprycel (dasatinib) and Tarigna (nilotinb), which also block the BCR-ABL cancer gene. Patients who have not had success with Gleevec are usually given Sprycel and Tarigna. All three drugs are taken orally. A bone marrow transplant is the only current way of curing a patient with CML. The younger the patient is the more likely the transplant will be successful.

Synribo (omacetaxine mepesuccinate) was approved by the FDA, on 26th October 2012, for the treatment of chronic myelogenous leukemia (CML) in adult patients who had been treated with at least two drugs, but whose cancer continued to progress. Resistance to medications is common in CML. Synribo is an alkaloid from Cephalotaxus harringtonia which inhibits proteins that trigger the development of cancerous cells. The drug is administered subcutaneously.

Patients with Chronic Lymphocytic Leukemia (CLL) may not receive any treatment for a long time after diagnosis. Those who do will normally be given chemotherapy or monoclonal antibody therapy. Some patients with CLL may benefit from allogeneic stem cell transplantation (bone marrow transplant).

Rabbit antibodies help Leukemia patients - scientists from Virginia Commonwealth University reported in the journal Bone Marrow Transplantation (July 2012 issue) that rabbit antibodies can improve survival and reduce the occurrence of relapses in patients with leukemia and myelodysplasia who are receiving a stem transplant from an unrelated donor.

Leukemia patients' own T-cells achieve remission for over two years - patients who were infused with their own T-cells after they had been genetically altered to fight cancer tumors stayed in full remission for over 24 months. Researchers from the Perelman School of Medicine at the University of Pennsylvania presented their findings at the American Society of Hematology's Annual Meeting and Exposition in December 2012. All those who took part in the human study had advanced cancers - ten of them had chronic lymphocytic leukemia, and two children had acute lymphoblastic leukemia.

All leukemia patients, regardless of what type they have or had, will need to be checked regularly by their doctors after the cancer has gone (in remission). They will undergo exams and blood tests. The doctors will occasionally test their bone marrow. As time passes and the patient continues to remain free of leukemia the doctor may decide to lengthen the intervals between tests.

Chronic Leukemia

Source(google.com.pk)
Chronic Leukemia Biography


Leukemia
Leukemia is a cancer of the blood and bone marrow in which the body produces abnormal white blood cells. This eMedTV article offers an overview of leukemia, including information about types of the disease, its symptoms, and its treatment.

Leukemia Symptoms
For people with leukemia, symptoms commonly include fevers, night sweats, frequent infections, and fatigue. This eMedTV article describes signs and symptoms of this disease, which may also include bruising easily, weight loss, and headaches.

Causes of Leukemia
The exact causes of leukemia are not yet known. As explained in this eMedTV article, however, researchers have identified risk factors (such as exposure to very high levels of radiation) that increase one's chances of developing leukemia.

AML
In AML, myeloblasts (leukemia cells) are abnormal and do not mature into healthy white blood cells. This eMedTV article describes this condition in detail, offering information on possible symptoms, treatment options, prognosis, and more.
Chronic Lymphocytic Leukemia
Chronic lymphocytic leukemia is type of cancer that occurs when too many white blood cells are produced. This eMedTV article offers an overview of CLL, with detailed information on risk factors, treatment options, prognosis, and more.

Childhood Leukemia
Childhood leukemia is a form of cancer in which blood-forming tissue produces abnormal blood cells. This eMedTV article describes the types of leukemia most common among children, including information about risk factors and symptoms.

Acute Leukemia
Acute leukemia is a cancer that starts in blood-forming tissue and progresses very quickly. This eMedTV resource takes an in-depth look at this condition, including possible causes, symptoms, treatment options, and more.

Cytoxan
A doctor may prescribe Cytoxan to treat various types of cancer, such as leukemia and breast cancer. This eMedTV resource lists other types of cancer that can be treated with Cytoxan, explains how the drug works, and offers dosing information.
Chronic Leukemia
Chronic leukemia is a slowly developing form of cancer that causes the production of abnormal blood cells. This eMedTV segment discusses types, causes, and symptoms of this form of leukemia, and describes tests used to diagnose the cancer.

Acute Lymphocytic Leukemia
Acute lymphocytic leukemia accounts for about 3,800 new cases of leukemia each year. This section of the eMedTV archives describes this type of leukemia, which is the most common type of leukemia in young children but can also affect adults.

Types of Leukemia
As this eMedTV page explains, the four most common types of leukemia include chronic lymphocytic leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, and acute myeloid leukemia. This article describes these types of the condition in detail.

Rituxan
Rituxan is a drug prescribed to treat rheumatoid arthritis, certain types of cancer, and other conditions. This eMedTV page offers an in-depth look at this medication, including how it works, side effects, dosing tips, and general safety precautions.

Chronic Myelogenous Leukemia
Chronic myelogenous leukemia is a disease in which the bone marrow produces too many white blood cells. This eMedTV article provides an overview this condition, with information on the causes, symptoms, treatment options, and prognosis.

Leukemia Cells
Leukemia cells are abnormal cells produced by blood-forming tissue. As this segment of the eMedTV Web site explains, there are two main types of these cells and they cause different symptoms and types of leukemia.

Hairy Cell Leukemia
Hairy cell leukemia occurs when cancer cells develop in the blood and bone marrow. This section of the eMedTV library provides an overview of this condition, including information about its symptoms, tests used to detect the disease, and treatment.

Early Symptoms of Leukemia
Early symptoms of leukemia may include fatigue, fever, and pain or a feeling of fullness below the ribs. This eMedTV Web page discusses early symptoms of leukemia in both acute and chronic cases of the cancer.
Gleevec
This eMedTV segment takes a detailed look at Gleevec, a chemotherapy drug used to treat leukemia, a rare skin cancer, and a type of tumor affecting the GI tract. This article offers information on how this drug works, side effects, dosing, and more.

Childhood Leukemia Symptoms
As this eMedTV Web page explains, symptoms of childhood leukemia commonly include fatigue, fever, and frequent infections. This article provides a list of symptoms occurring with acute lymphoblastic leukemia (ALL) and acute myelogenous leukemia (AML).

Leukemia Treatment
As this eMedTV page explains, treatment options for leukemia may include chemotherapy, radiation therapy, and surgery. This article discusses these and other treatments, and includes information about side effects, second opinions, and clinical trials.

Leukemia Statistics
Based on leukemia statistics, 35,070 people will be diagnosed with the disease in the United States in 2006. This eMedTV page contains various statistics on this disease, including survival rates, age-at-diagnosis figures, and lifetime risk percentages.

Myeloid Leukemia

Source(google.com.pk)
Myeloid Leukemia Biography

Nora Ephron's final act played out in Manhattan on June 26 where the 71-year-old writer and movie director died from pneumonia brought on by acute myeloid leukemia (AML), one of the most common types of leukemia among adults. AML is a cancer caused when abnormal cells grow inside bone marrow and interfere with the production of healthy blood cells. The marrow eventually stops working correctly, leaving a person with an increased risk of bleeding and infections.

Ephron—best known for writing When Harry Met Sally and Sleepless in Seattle—was first diagnosed in 2006 with one of the myelodysplastic syndromes (MDS), a category of blood diseases also referred to as "preleukemia" that can progress into AML if the bone marrow continually fails to produce enough healthy platelets, red blood cells and white blood cells over time. MDS made headlines recently when ABC's Good Morning America anchor Robin Roberts announced she has been diagnosed with the disease.

Some types of leukemia, including AML, develop as a result of exposure to certain chemicals (including herbicides and pesticides), chemotherapy drugs (such as etoposide and a class of drugs known as alkylating agents) and radiation. Typically, however, a doctor is unable to pinpoint the exact cause in individual cases.

Although estimates vary, there are between 10,000 and 12,000 new cases of MDS in the U.S. annually. More than 80 percent of all MDS patients are older than 60. The National Cancer Institute projects that 13,780 men and women—7,350 men and 6,430 women—will be diagnosed with AML and that 10,200 men and women will die of the malady this year.

Scientific American spoke with Bart Scott, a medical oncologist specializing in the treatment of patients with MDS, about syndrome's progression to AML, who is most at risk for this cancer and whether there are any promising treatments on the horizon. Scott is also director of hematology and hematologic malignancies at the Seattle Cancer Care Alliance and an assistant member of the Fred Hutchinson Cancer Research Center's clinical research division.

What is the typical trajectory for this form of cancer?
The trajectory of myelodysplastic syndrome is highly dependent upon two factors—the bone marrow myeloblast count and the cytogenetics. [A myeloblast is an immature blood cell that will eventually develop into a type of white blood cell.] The higher the bone marrow myeloblast count is at time of diagnosis the more likely a patient is to progress to acute myeloid leukemia. In general, the more abnormal the cytogenetics [a cell's hereditary and functionalm characteristics] are the more likely a patient is to progress to AML, but there are certain abnormalities that have a good prognosis.

Who are the typical victims of this form of leukemia?
The median age of diagnosis of MDS is 72, it is slightly more common in men than women, and there is a correlation between incidence and age. I would say that Ephron's course does seem consistent with MDS progression to AML.

What are the risk factors for this cancer?
They are based on increasing age as well as certain environmental exposures like pesticides and herbicides. This is not casual contact—we are talking about prolonged, persistent exposure. The amount of exposure required to cause the disease is unknown, however.

Given that Ephron lived most of her life in New York City, whose residents are not typically exposed to inordinately large amounts of pesticides or herbicides, what might account for her condition?
Some researchers have tried to understand MDS in terms of clustering, although there is very little clustering data available. The idea is to understand whether MDS tends to cluster in areas of higher exposure to certain toxic agents. In 2007 Xiaomei Ma [an associate professor of epidemiology the Yale School of Public Health] and a team of researchers published a paper in Leukemia Research regarding this phenomenon. They found that cases of MDS in Connecticut were clustered near the western border. In general, clustering supports the idea that environmental exposure is contributory to the development of a disease. This is the only paper that I know of that has demonstrated clustering in MDS.

We are doing a similar study in the state of Washington. Hanford Site is a nuclear production complex near the Tri-Cities area in southeastern Washington [encompassing Kennewick, Pasco and Richland]. We are looking to see if there is clustering in the Tri-Cities area. But these studies are difficult given the mobility of the area's population, unknown exposure time and any delay in the development of MDS after exposure.

What is the usual treatment?
There are three FDA approved treatments. One is azacitidine, approved by the FDA in 2004 and marketed as Vidaza. Decitabine, sold as Dacogen and approved in 2010 to treat MDS, is another option. [The FDA's oncologic drugs advisory panel in February, however, recommended against the approval of Dacogen to treat older patients with AML. The drug's maker wants FDA approval to use Dacogen in AML patients 65 years and older and who are not considered good candidates for high-dose chemotherapy as an initial treatment for AML.] The third is lenalidomide, also known as Revlimid and introduced in 2004.

However, the only curative treatment is stem cell transplantation. Stem cells are infused just like a blood transfusion. Sources of stem cells include cord blood cells, peripheral blood mobilized stem cells or bone marrow. Success depends on the stage of disease. The one-year treatment related mortality is approximately 20 percent. If patients are transplanted in an early stage of the disease, we have a success rate of 80 percent.

Are there any promising treatments in late-stage testing?
Yes, there are several drugs being investigated in phase II and phase III trials. We currently have a phase III trial open with a drug called rigosertib for patients who have failed prior treatment with azacitidine or decitabine, which are known as demethylating agents. The proposed mechanism of action for these drugs is that they alter gene expression profiles in the cancer cells and increase their susceptibility to death. Rigosertib is a cell cycle inhibitor and would prevent the cancer cells from growing and induce direct damage to the cancer cells, causing their death.

Cancer In The Blood

Source(google.com.pk)
Cancer In The Blood Biography

Cancer is the leading cause of disease-related deaths in children, and leukemia is the most common form of childhood cancer.1, 2 Leukemia survival rates have improved significantly in the last three decades as treatment protocols have been optimized. However, this success is associated with a two to fourfold increased rate of adverse late effects of therapy.3, 4 Furthermore, children with T-cell acute lymphoblastic leukemia (T-ALL) who experience a bone marrow relapse have a particularly poor prognosis,5, 6 and currently the best therapeutic option is hematopoietic stem cell transplantation. Unfortunately, induction of complete remission, a necessary first step for transplantation, is extremely difficult in relapsed T-ALL.7 Consequently, there continues to be a need for new therapeutic strategies, such as molecularly targeted therapies, that can interact synergistically with current treatment protocols and result in improved efficacy and reduced side effects.

Mer receptor tyrosine kinase (MerTK), a member of the TAM (Tyro3/Axl/Mer) family of receptor tyrosine kinases, was initially cloned from a B-lymphoblastoid complementary DNA library.8 In normal hematopoiesis, MerTK is important in macrophage and dendritic cell inflammatory response and MerTK promotes phagocytosis of apoptotic cells. Other physiological roles for MerTK in hematopoietic cells, include natural killer-cell differentiation9 and platelet aggregation.10 MerTK is activated by the ligands Protein S and Gas6, two structurally similar vitamin K-dependent proteins.11, 12 Both ligands are produced by a variety of tissues and are present in serum.13, 14 Interestingly, overexpression of both Gas6 and Protein S is correlated with poor prognosis in a variety of cancers.15 More recently, Tubby, tubby-like protein 1 and galectin-3 have been described as novel Mer ligands important in phagocytosis.16, 17 Although galectin-3 has been shown to be important in tumorigenesis,18 it is unclear whether Tubby or tubby-like protein 1 have a role in cancer.

The oncogenic potential of MerTK is supported by various lines of evidence. MerTK activation results in upregulation of the MAPK and PI3K/Akt antiapoptotic signaling pathways, thereby promoting survival of tumor cells. Expression of activated MerTK is transforming in Ba/F3 pro-B lymphocytes and NIH 3T3 fibroblasts;19, 20 MerTK overexpression has been reported in a variety of human cancers, including B- and T-ALL21, 22 and ectopic expression of MerTK in lymphocytes in a transgenic mouse model promotes the development of leukemia/lymphoma.23 Additionally, our lab and others have shown that MerTK inhibition leads to increased sensitivity of glioblastoma24 and non small-cell lung cancer 25 cell lines to treatment with chemotherapeutic agents. Taken together, these data indicate important roles for MerTK in leukemogenesis and chemoresistance.

Here, we extend our previous work on MerTK expression in T-ALL.21 We show increased expression of MerTK in a prospective analysis of pediatric T-cell ALL at the time of diagnosis. Furthermore, we show that MerTK stimulation leads to activation of the MAPK pathway and the signal transducers and activators of transcription (STAT) proteins, two prosurvival signaling pathways. Lastly, we demonstrate that MerTK inhibition sensitizes leukemia cells to treatment with chemotherapeutic drugs, and reduces their leukemogenic potential in a xenograft mouse model of leukemia. These data support the development of MerTK-targeted therapies for the treatment of pediatric T-ALL.

Materials And Methods

Patient Samples And Cell Culture
Diagnostic bone marrow or peripheral blood samples were obtained at the Children’s Hospital Colorado (Aurora, CO, USA) according to an institutionally approved COMIRB protocol. The CEM, HSB-2, Jurkat, Loucy, Molt3 and Molt4 cell lines were obtained from the American Type Culture Collection (Manassas, VA, USA). The ALL-SIL, DND41, HPB-ALL, SupT11 and TALL-1 cell lines were obtained from the German Collection of Microorganisms and Cell Cultures (Braunschweig, Lower Saxony, Germany). All cell lines were maintained in RPMI medium (Hyclone Laboratories, Logan, UT, USA) supplemented with 10% heat inactivated fetal bovine serum (Atlanta Biologicals, Lawrenceville, GA, USA) and penicillin (100 Units/ml)/streptomycin (100 μg/ml) (Hyclone Laboratories, Logan, UT, USA). The identity of all cell lines was confirmed by genomic fingerprinting via short tandem repeat microsatellite loci analysis.

Flow Cytometric Analysis Of Cell Surface Proteins
Flow cytometric analysis of cell lines was performed as previously described.26 For flow cytometric analysis of T-ALL patient samples, cells were stained with mouse anti-human Mer 590,27 biotinylated anti-human Axl (R&D Systems, Minneapolis, MN, USA), ECD-linked anti-human CD45 (Beckman Coulter, Brea, CA, USA) and PC7-linked anti-human CD7 (Beckman Coulter). PE-linked donkey anti-mouse (Jackson ImmunoResearch Laboratories, West Grove, PA, USA) and FITC-linked Streptavidin (BD Biosciences, San Jose, CA, USA) were used to detect anti-hMer and anti-hAxl antibodies, respectively. Stained cells were washed and resuspended in phosphate-buffered saline and analyzed using a FC 500 flow cytometer (Beckman Coulter) and CXP data analysis software (R&D Systems). Mer and Axl expression was determined on live, CD45/CD7 double-positive cells, with 20% expression considered positive.

Western Blot Analysis
Whole-cell lysates were prepared in lysis buffer (50 mM HEPES pH 7.5, 150 mM NaCl, 10 mM EDTA, 10% glycerol and 1% Triton X-100) supplemented with phosphatase inhibitors (0.1 mM Sodium pervanadate and 0.1 mM Na2MoO4) and protease inhibitors (Complete mini, Roche Molecular Biochemicals, Indianapolis, IN, USA). Sodium pervanadate was prepared per the Alliance for Cellular Signaling (http://www.signaling-gateway.org/) protocol. Samples were analyzed by SDS−polyacrylamide gel electrophoresis and immunoblotting with the following primary antibodies: anti-Actin (sc-1616, Santa Cruz Biotechnology, Santa Cruz, CA, USA); anti-phospho-Mer (pMer Y749/Y753/Y754, PhosphoSolutions, Aurora, CO, USA); anti-hMer (no. 1633-1, Epitomics, Burlingame, CA, USA); anti-phospho-p44/42 MAPK (Thr202/Tyr204, no. 9106), anti-p44/42 MAPK (no. 9102), anti-PARP (no. 9542), anti-Caspase 3 (no. 9662), anti-phospho-Stat5 (Y694, no. 9359), anti-Stat5 (no. 9358) (Cell Signaling Technology, Danvers, MA, USA). Primary antibodies were labeled with horseradish peroxidase-conjugated secondary antibodies (donkey-anti-goat, sc-2020, Santa Cruz Biotechnology; goat-anti-mouse, 170–6516, Bio-Rad Laboratories, Hercules, CA, USA; or donkey-anti-rabbit, 711-035-152, Jackson ImmunoResearch Laboratories, West Grove, PA, USA) and proteins were visualized by enhanced chemiluminescence (Perkin-Elmer, Waltham, MA, USA).

Human Phospho-Kinase Array Analysis
Jurkat cells were serum starved for 3 h, and then separate aliquots were treated with vehicle control or 200 nM rhGas6 (R&D Systems) for ten minutes. Cell lysate isolation and the phospho-array screen were conducted according to the Human Phospho-Kinase Array Kit (R&D Systems) protocol.

Lentiviral Small Hairpin RNA (shRNA) Vectors, Mer Expression Vector, luciferase Vector And Target Cell Transduction
Lentiviral shRNA vectors shMer1 (targeting Mer) and shControl1 (targeting GFP) have been previously described.26 The lentiviral shRNA vector shControl2 (SHC002, Sigma, St. Louis, MO, USA) expresses an shRNA construct that does not target any human gene. shRNA expressing Jurkat cell lines have been previously described.26 shRNA expressing HSB2 cell lines were derived in the same manner as with the Jurkat cell lines. The identity of all shRNA expressing cell lines was confirmed by genomic fingerprinting. The construction of the Mer expression vector and generation of the stable Jurkat Mer add-back cell lines has been previously described.26 Details of luciferase tagging of Jurkat parental and shRNA derivative cell lines will be provided elsewhere.

Cellular Metabolism Assay
Initial experiments were performed to determine the maximum number of cells that could be plated while still remaining within the linear range of the colorimetric assay. Jurkat and HSB2 (both at 5 × 105 cells/ml) were plated in 96-well plates and cultured for 4 h or overnight, respectively. Chemotherapeutic agents or vehicle only were added at the appropriate concentrations and the cells were cultured for an additional 48 h. MTT (3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide, Sigma) reagent was added to the cells to a final concentration of 0.65 mg/ml for the final 3 h of culture. Solubilization solution (2X, 10% SDS in 0.01M HCl) was added overnight and optical density was determined at 562 nm with a reference wavelength of 650 nm. Relative cell numbers were calculated by subtraction of background absorbance and normalization to untreated controls. IC50 values were determined by nonlinear regression of the MTT assay data using Prism (Version 5.0, GraphPad Software, LaJolla, CA, USA).

Flow Cytometric Analysis Of Apoptosis
Jurkat or HSB2 parental and shRNA derivative cells were plated at 5 × 105 cells/ml and cultured for 48 h in the presence of the indicated agents or vehicle control. Cells were collected by centrifugation (240 × g for 5 min), resuspended in phosphate-buffered saline containing 1 μM YO-PRO-1 (Invitrogen, Grand Island, NY, USA) and 1.5 μM propidium iodide (PI) (Invitrogen), and incubated on ice for 20–30 min. Fluorescence was detected and analyzed using a FC 500 flow cytometer and CXP data analysis software (Beckman Coulter).

Methylcellulose Assay
Five hundred Jurkat parental and shRNA derivative cells were plated in methylcellulose-based medium (Cat no. 1101, Reachbio, Seattle, WA, USA) according to manufacturer’s protocol. Cells were grown for 8 days, then stained with MTT (0.5 mg/ml) overnight and visualized with a colony counter (GelCount, Oxford Optronix, Oxford, UK). Colonies greater than 50 μm in diameter were counted.
Xenograft model Of leukemia And In Vivo Iuciferase Imaging
NOD scid gamma mice (Stock no. 5557, The Jackson Laboratory, Bar Harbor, ME, USA) were irradiated with 200 rads and injected intravenously with Jurkat or shRNA expressing cell lines (5 × 106 cells). Animals were monitored daily and sacrificed upon signs of leukemia onset (weight loss, decreased activity and/or hind limb paralysis). Whole blood, bone marrow and spleen were harvested at the time of sacrifice and analyzed for human cell-surface proteins CD2 and CD45 by flow cytometry. Mer surface expression was confirmed by flow cytometry and western blot. Control animals injected with phosphate-buffered saline did not develop leukemia. For luciferase imaging of leukemia progression, monoclonal Jurkat and shRNA-derivative cell lines expressing similar levels of luciferase were generated using a lentiviral system which will be described elsewhere. Mice inoculated with luciferase-expressing cell lines were injected with K+Salt D-Luciferin (Caliper Life Sciences, Hopkinton, MA, USA) and imaged with a Xenogen IVIS200 (Caliper Life Sciences, Hopkinton, MA, USA) imager. All experiments involving animals followed the regulatory standards approved by the University of Colorado Institutional Animal Care and Use Committee.

Statistical analysis
For determination of IC50 values, experiments were repeated 4–12 times. When the variability within a cell line was approximately the same among all of the cell lines being compared, repeated analysis of variance was used to estimate and compare the mean IC50 values. Otherwise, the difference in IC50 between cell lines was calculated and a t-test was used to determine if the mean difference was significantly different from 0. SAS 9.2 software (SAS Institute Inc., Cary, NC, USA. SAS/STAT 9.2) was used for the analyses. All other statistical analyses was carried out using Prism software (Version 5.0, GraphPad Software, LaJolla, CA, USA) Results were considered significant at P <0.05.

Results
Mer Receptor Tyrosine Kinase Is Expressed In Pediatric T-ALL
We have previously shown that MerTK is ectopically expressed in pediatric T-ALL patient samples using a retrospective analysis of banked T-cell leukemia patient samples (21). In this report, we expand on that observation by analyzing MerTK expression in a panel of eleven T-ALL cell lines. Western blot analysis of protein extract from each of the cell lines showed that 6 out of the 11 lines (54.5%) expressed MerTK protein at varying levels, with DND41 expressing the least and Loucy expressing the most (Figure 1a). The results of the western blot analysis and the relative levels of MerTK protein expression were also confirmed by flow cytometric analysis of surface protein expression (data not shown). The observed variability in MerTK expression levels is consistent with reports in other tumor types, including gliobastoma and non small-cell lung cancer, where negative as well as low and high MerTK expressing cell lines have been identified.23, 25
Figure 1.

MerTK expression in T-ALL cell lines and diagnostic patient samples. (a) MerTK expression in a panel of T-ALL cell lines was detected by western blot analysis. (b) MerTK expression in diagnostic pediatric T-ALL patient samples was detected by flow cytometry analysis. Expression was determined after gating on live CD45 and CD7 positive lymphoblasts.

Full figure and legend (85K)

MerTK expression was also analyzed prospectively in a set of diagnostic samples obtained at the time of patient admission (see Table 1). Upon determination of the T-ALL diagnosis, CD45/CD7 double positive cells were tested for MerTK and Axl expression using flow cytometry (Figure 1b). As shown in Table 1, 5 out of 12 patient samples (41.7%) analyzed in the Children’s Hospital Colorado clinical pathology laboratory were classified as positive for MerTK expression, based on the established clinical criteria that at least 20% of cells must express a specific marker to be considered positive. As shown in Figure 1b, in MerTK positive samples the fluorescence intensity of the entire population shifted along the MerTK expression axis, suggesting that MerTK expression was increased in all cells in patient samples and was not solely due to the presence of a MerTK-expressing subclone. These prospective findings in patient samples, as well as the data for cell lines, are consistent with the findings previously reported by our lab, which showed MerTK mRNA expression in 19/34 (55.8%) banked patient samples and MerTK protein expression in 8/16 (50%) banked leukemia patient samples.21

Multiple signaling pathways are activated upon Gas6 stimulation of Mer receptor tyrosine kinase
Previous reports have shown that a variety of signaling pathways are activated downstream of Gas6/MerTK signaling (reviewed in28, 29). We first analyzed the status of MerTK activation in the Jurkat and HSB2 cell lines and the ability of the receptor to respond to Gas6 stimulation (Figure 2a). MerTK was phosphorylated in both the Jurkat and HSB2 cell lines in response to Gas6 treatment. Additionally, in the Jurkat cell line there was low but detectable constitutive activation of MerTK, which was not present in the HSB2 cell line. To identify the pathways that might specifically be involved in the role of MerTK in T-ALL we used a human phospho-kinase array to analyze extracts from Jurkat cells treated with Gas6 or vehicle only (Figure 2b). As expected, we detected an increase in phosphorylated ERK and AKT in the Gas6 treated sample indicating the activation of the MAPK and PI3K/AKT pathways. Interestingly, an increase in pSTAT5 and pSTAT6 was also noted (Figure 2b), suggesting the activation of the Stat pathway in a MerTK dependent manner.
Figure 2.

MAP Kinase and Stat pathway activation in response to MerTK Gas6 stimulation. (a) Analysis of phosphorylation status of MerTK in Jurkat and HSB2 cell lines during log growth (lane 1), or incubated in serum-free media for 3 h and then treated with vehicle control (lane 2) or Gas6 (200 nM) (lane 3). (b) Human phospho-kinase array analysis of the Jurkat cell line treated with Ga6 (200 nM) or vehicle control. (c and d) Western blot analysis of ERK and Stat5 activation in response to Gas6 (200 nM) treatment in (c) Jurkat and (d) HSB2 parental and shRNA derivative cell lines.

The results from the phospho-array screen were verified by western blot analysis in the T-ALL cell lines Jurkat and HSB2. Additionally, to confirm the requirement of MerTK in the Gas6 induced signaling, we derived cell lines where MerTK expression is inhibited by an shRNA construct (shMer1) targeting the 3′ UTR of the MerTK transcript. As shown in Figures 2c and d, MerTK protein expression was essentially ablated in two independent clones of Jurkat and HSB2 expressing shRNA directed against MerTK (shMer1A and shMer1B), while the cell lines expressing non-silencing control shRNA constructs (Jurkat shCntrl1 and HSB2 shCntrl2) showed no change in the level of MerTK protein.

Treatment of Jurkat and HSB2 parental and shControl cell lines with Gas6 led to an increase in the phosphorylation of Erk 1/2 (Thr202/Tyr204), whereas in the MerTK knockdown cell lines the activation of Erk was attenuated (Figure 2c and d), indicating that MerTK is required for Erk signaling in these cell lines. Similarly, when we analyzed the phosphorylation status of Stat5 in the Jurkat cell lines, both the Jurkat parental and shCntrl1 cell lines showed an increase in phosphorylated Stat5 (Tyr694) (pStat5), and this signal was absent in the Jurkat MerTK knockdown cell lines (Figure 2c).

In contrast to the Jurkat cell lines, the HSB2 parental and shCntrl2 cell lines showed a significant basal level of pStat5 (Figure 2d), which was largely unresponsive to treatment with Gas6. Interestingly, in the HSB2 MerTK knockdown cell lines, basal levels of pStat5 were reduced compared with the controls. However, when the total levels of Stat5 protein were analyzed, the knockdown cell lines showed a lower level of total Stat5 than in the HSB2 parental or shCntrl2 cell lines. These findings suggest that MerTK may have a role in the regulation of Stat5 activation and expression.

Inhibition of MerTK Increased The Chemosensitivity Of T-ALL Cell Lines
As MerTK stimulation leads to the activation of Erk 1/2 (Figure 2) and other prosurvival pathways,19, 30 we decided to determine the effect of MerTK inhibition on the response of the Jurkat and HSB2 cell lines to treatment with cytotoxic agents that are currently used clinically.31 Using the MTT assay, the IC50 of the HSB2 parental and shRNA derivative cell lines in response to treatment with Cytarabine (Ara-C), Etoposide or 6-mercaptopurine (6-MP) were determined; similarly, the response of the Jurkat cell lines to treatment with Methotrexate was also analyzed.
As can be seen in Figures 3a, b and c), inhibition of MerTK increased the sensitivity of HSB2 to Ara-C, Etoposide and 6-MP. Comparison of the IC50 values (Table 2), generated by nonlinear regression of the MTT assay data, showed that the HSB2 shMer knockdown cell lines were significantly more sensitive to Etoposide (IC50=20.5 and 25.8 nM) compared with HSB2 shCntrl2 cells (IC50=44.1 nM; P=0.014 and 0.047 vs HSB2 shMer1A and shMer1B, respectively). In response to 6-MP, there was a statistically significant ~1.8-fold increase in the sensitivity of the HSB2 shMer cells (IC50=2.78 and 2.63 μM) in comparison with the HSB2 shCntrl2 cells (4.79 μM; P=0.019 and 0.033 vs HSB2 shMer1A and shMer1B, respectively). In response to treatment with Ara-C, there was a ~fourfold decrease in the IC50 of the HSB2 shMer cells (7.5 and 9.8 nM), relative to shCntrl2 cells (39.3 nM); however, this decrease was not statistically significant (P=0.125 and 0.162 vs HSB2 shMer1A and shMer1B, respectively). We saw a similar result when the Jurkat cell lines were tested in response to treatment with Methotrexate (Figure 3d), with both Jurkat shMer knockdown lines (IC50=44.8 and 43 nM) significantly more sensitive than the Jurkat shCntrl cell line (IC50=64.3 nM; P=0.005 and 0.033 vs Jurkat shMer1A and shMer1B, respectively) (Table 2).

Inhibition of MerTK leads to increased sensitivity of HSB2 and Jurkat cell lines to chemotherapeutic agents. MTT analysis of HSB2 and Jurkat parental and shRNA derivative cell lines in response to increased concentrations of chemotherapeutic agents. (a, b, c) Analysis of HSB2 treated with Ara-C, Etoposide or 6-MP, respectively. (d) Analysis of Jurkat treated with Methotrexate. Error bars represent standard error of the mean derived from at least four independent experiments.

The observed changes in chemosensitivity of the shMer knockdown cell lines in the MTT analysis can be attributed to either a decrease in proliferation or an increase in cell death. To determine whether decreased Mer expression enhanced cell death in response to treatment with chemotherapeutic agents, the levels of apoptosis were analyzed with a fluorescence-based assay, using YOPRO-1 and PI to stain apoptotic and dead cells, respectively. As shown in (Figure 4a), both HSB2 shMer knockdown cell lines had a statistically significant increase in the percentage of dead cells in response to treatment with 20 nM Ara-C (70.8 and 44%) or 6-MP (52–68.5 and 32.9–54.6%), compared with the HSB2 shCntrl2 cell line (21% for 20 nM Ara-C and 16.5–35.9% for 6-MP, P<0.001). Similarly, when Jurkat cells were treated with 60 nM Methotrexate (Figure 4b), there was a significantly increased level of apoptotic cells in the shMer knockdown cell lines (31.9 and 30.7%) compared with the Jurkat shCntrl1 cell line (13%; P<0.01). Analysis of the results from the YOPRO/PI assay using the Bliss independence model demonstrated an additive or synergistic increase in sensitivity to chemotherapy treatment in both the HSB2 and Jurkat cell lines depending on the concentrations of chemotherapy used (Supplementary Table 1).

Inhibiton of MerTK leads to increased apoptosis in HSB2 and Jurkat cell lines in response to treatment with chemotherapeutic agents. (a and b) Flow cytometry analysis of apoptotic and dead cells stained with YOPRO and PI. Error bars represent s.e. of the mean of at least three independent experiments. Statistical comparison vs shControl by two-way repeated measures analysis of variance followed by Bonferroni posttests(**P<0.01, ***P<0.001) (c and d) western blot analysis of Caspase 3 activation and PARP cleavage in (c) HSB2 and (d) Jurkat parental and shRNA derivative cell lines in response to Ara-C or Methotrexate (MTX) treatment, respectively.


To confirm the results from the YOPRO/PI analysis on a molecular level, the status of the apoptosis signaling effectors Caspase 3 and PARP were analyzed. As shown in Figure 4c, when HSB2 shMer cells were treated with Ara-C, there was an increase in cleaved Caspase 3 and cleaved PARP compared with the parental and shCntrl2 cell lines, consistent with the increased apoptosis observed in the YOPRO/PI assay (Figure 4a). Similar results were observed in the Jurkat cell lines (Figure 4d), with both the Jurkat shMer cell lines showing increased activation of Caspase 3 and PARP cleavage when treated with Methotrexate relative to the Jurkat parental and shCntrl1 cell lines.

Inhibition of MerTK Decreases The Oncogenic Potential Of The Jurkat Cell Line
To determine whether MerTK inhibition resulted in any changes in the clonogenic potential of T-ALL, the colony forming potential of the Jurkat parental and shRNA derived cell lines in methylcellulose media was analyzed. As shown in Figure 5a there was a significant decrease in the ability of the shMer knockdown cell lines to form colonies (75 and 67colonies) relative to the shCntrl1 cell line (185 colonies, P<0.01), indicating that MerTK activity contributes to the clonogenic potential of the Jurkat cell line. The colony formation assay was not carried out with the HSB2 cell line, as the cells did not form distinct colonies in this assay.


Inhibition of MerTK impairs the clonogenic potential of the Jurkat cell line. (a) Jurkat parental and shRNA derivative cells were plated in methylcellulose media, grown for 8 days, and colonies were counted. (b) Analysis of methylcellulose colony formation in Jurkat parental and shRNA derivative cells transduced with an empty (pLNCX) or a MerTK expression vector. Error bars represent the s.e. of the mean of at least three independent experiments. Statistical comparison vs shCntrl1 by one-way repeated measures analysis of variance followed by Bonferroni posttests. The differences in colony number between the Jurkat parental, shCntrl1 and shMer1A+MerWT lines were not statistically significant.

In order to control for the possibility of off-target effects of the shRNA used for Mer knockdown, we generated a MerTK add-back cell line derived from the Jurkat shMer1A cell line. As is shown in Figure 5b, reintroduction of MerTK expression in the knockdown cell line restores the clonogenic potential of the cells to the same level as the Jurkat parental and shCntrl1 cell lines with the empty vector only. Additionally, the increased chemosensitivity in the knockdown cell lines is reversed by exogenous MerTK expression (Supplementary Figure 1), suggesting that the phenotypes observed as a result of shRNA-mediated knockdown are not due to off-target effects and are specific to MerTK inhibition.

To further investigate the effect of MerTK on the oncogenic potential in T-ALL, a mouse xenograft model of leukemia was developed by transplating Jurkat parental and shRNA-derived cell lines into NOD scid gamma mice. A significant survival advantage was observed when mice were injected with Jurkat shMer cell lines, compared with those injected with Jurkat shCntrl1 (P<0.0001) (Figure 6a). Kaplan−Meier survival curves were used to determine a media survival of 60 and 59 days for mice injected Jurkat shMer1A and shMer1B, respectively, compared with a median survival of 30.5 days for mice injected with the Jurkat shCntrl1 cell line. Interestingly, a slight difference in survival was also noted between mice injected with the control cell line compared with those injected with the Jurkat parental cell line (27 vs 30.5 days).

Inhibition of MerTK significantly improves survival and delays disease progression in a Jurkat induced xenograft model of human leukemia. (a) Kaplan−Meier survival curves derived from NOD scid gamma mice mice injected with Jurkat parental or shRNA derivative cell lines. (b) Luciferase imaging of leukemia progression in NOD scid gamma mice mice injected with luciferase-tagged Jurkat and shRNA derivative cell lines and luciferin.

In order to visualize the extent of the changes in leukemia onset and progression in a more dynamic manner, mice were transplanted with monoclonal Jurkat cell lines (parental, shControl and shMer) expressing luciferase and luciferase activity was visualized over time as leukemia developed. Consistent with the data from the survival curve, mice injected with the parental cell line showed a slightly greater leukemia burden earlier on relative to the shCntrl1-injected mice (Figure 6b). Furthermore, we are able to visually observe the significant difference in survival induced by MerTK knockdown, as the mice that were injected with the Jurkat shMer1A/Luc cell line had dramatically less leukemia burden at days 17 and 24 postinjection relative to either the parental or shCntrl1 cell lines.

Discussion
In this paper, we have extended our previous findings demonstrating ectopic expression of MerTK in pediatric T-cell ALL.21 We detected MerTK expression in 5 out of 12 (41.7%) pediatric patient samples at the time of diagnosis, and in 6 out of 11 (54.5%) T-ALL cell lines analyzed. These findings are consistent with the data previously reported (MerTK expression detected in 50–55.8% of banked pediatric T-ALL patient samples),21 and also demonstrate the relative ease with which MerTK expression analysis can be incorporated into current flow cytometry diagnostic protocols.

To better understand the mechanism by which MerTK activation provides an advantage in T-cell leukemia cells, we screened a human phospho-kinase array for changes in phosphorylation status of proteins in response to Gas6-mediated activation of MerTK. Of the 38 different phospho-proteins present on the array, six proteins exhibited a change in phosphorylation status, including the ERK1/2 and AKT kinases and the STAT5 protein. Activation of the STAT pathway has been observed in AML and ALL, and is known to occur through a variety of mechanisms, including expression of Jak kinase fusion proteins (for example, TEL-JAK2).32 Although there are a few reports of STAT activation downstream of the TAM (Tyro3/Axl/Mer) receptors,33 MerTK-dependent STAT phosphorylation has only been observed in COS cells overexpressing a constitutively active chimeric MerTK receptor.34 In this report, we show that Stat5 is phosphorylated in response to Gas6 treatment and that MerTK inhibition reduces this response. Our data are the first to demonstrate a ligand-dependent role for endogenous MerTK protein in STAT signaling. In addition, STAT signaling as a consequence of MerTK activation has not been previously reported in leukemia. The mechanism of MerTK-dependent Stat5 phosphorylation will require further study, as we were not able to detect changes in Jak kinase activation (data not shown). It is possible that Stat5 phosphorylation could be mediated by another kinase, such as Src or MAP kinases,35 which have also been shown to be activated in response to MerTK stimulation.

Both the ERK1/2 and AKT kinases are well known modulators of antiapoptotic signals in cancer cells and have been shown to be downstream of MerTK stimulation. Both pathways can be activated by a variety of inputs and mechanisms. For example, in Jurkat cells AKT is constitutively active due to a mutation in PTEN, a phosphatase that regulates AKT activity.36 Thus, AKT activity in Jurkat cells was not significantly affected by MerTK stimulation or inhibition (data not shown). ERK1/2 activation, however, was responsive to Gas6 treatment in the Jurkat and HSB2 cells, and was blunted in the shRNA derived Mer knockdown cell lines.

The results of our signaling analysis suggests that targeted inhibition of MerTK would reduce prosurvival signaling in cells and result in a more effective outcome in response to cytotoxic treatment. Accordingly, both Jurkat and HSB2 MerTK knockdown cells were more sensitive to treatment with chemotherapeutic agents currently used for the treatment of leukemia.31 The increased sensitivity was mediated by a higher rate of apoptosis, which was detected by increased levels of caspase 3 and PARP cleavage. Interestingly, MerTK inhibition also reduced the clonogenic potential of the Jurkat cell line, both in an in vitro assay and in a mouse model of leukemia. Importantly, we were able to rescue the increased chemosensitivity and reduced clonogenic potential in the MerTK knockdown cell lines by exogenous expression of MerTK, indicating that the results presented here are unlikely to be due to off-target effects of the shRNA construct.

In conclusion, in this report we have confirmed that MerTK is ectopically expressed in ~50% of patients with T-cell ALL, and we demonstrated that MerTK expression analysis can be incorporated into current flow cytometry diagnostic protocols. We have shown that MerTK activation leads to upregulation of prosurvival signaling pathways in T-cell lines, and identified novel downstream signaling via Stat5. Furthermore, we have shown that inhibition of MerTK increases the sensitivity of leukemia cells to treatment with cytotoxic agents, decreases their colony-forming potential, and MerTK inhibition also decreased the leukemogenic potential of T-cell lines in a mouse model. We believe these results offer compelling evidence that MerTK is a viable target for the development of targeted inhibitors that can be used in combination with cytotoxic therapies for T-cell leukemia, and that these inhibitors will increase the benefit of current treatment protocols while allowing for dose reduction and a decrease in the severity of the side effects that are now observed.

What Is Blood Cancer


Source(google.com.pk)
What Is Blood Cancer Biography
Blood cancer is a form of cancer which attacks the blood, bone marrow, or lymphatic system. There are three kinds of blood cancer: leukemia, lymphoma, and multiple myeloma. These malignancies have varying prognoses, depending on the patient and the specifics of the condition, but overall survival rates with blood cancer increased radically in the late 20th century with the development of advanced treatments. When caught early, blood cancer can be very manageable in some cases, which is one very good reason to make regular trips to the doctor a priority for people of all ages.

In the case of leukemia, the cancer interferes with the body's ability to make blood. Leukemia attacks the bone marrow and the blood itself, causing fatigue, anemia, weakness, and bone pain. It is diagnosed with a blood test in which specific types of blood cells are counted. Treatment for leukemia usually includes chemotherapy and radiation to kill the cancer, and in some cases measures like bone marrow transplants may be required. There are several different types of leukemia, including chronic myelogenous leukemia, acute lymphoblastic leukemia, and hairy cell leukemia.

Swiss API quality at Asian prices. www.dolder.com/bendamustine
Lymphomas are blood cancers which involve the lymphocytes, a type of white blood cell. They are divided into Hodgkin's and non-Hodgkin's types. Lymphoma often involves swollen lymph nodes in addition to the symptoms for leukemia listed above, and it is also treated with chemotherapy and radiation. Multiple myeloma is a type of blood cancer which primarily appears in older people, involving the plasma, another type of white blood cell. Chemotherapy, radiation, and other drug treatments can be used to manage multiple myeloma.

The goal in treating blood cancer is to achieve remission, a situation characterized by the absence of symptoms. Even in remission, a blood cancer can still start up again, so people who have experienced blood cancer may need to attend regular follow-up medical appointments and annual checkups to check for a recurrence of the cancer. Blood cancer does not appear to be preventable, but like other cancers, the risk seems to be reduced among people who eat a healthy diet, exercise, and maintain good mental health.

People diagnosed with blood cancer can work with an oncologist, a doctor who specializes in cancer, or a hematologist, a doctor who specializes in diseases of the blood. Some patients work with both, attempting to develop a treatment plan which will be as effective as possible. Because individual cases can be quite varied, patients often benefit from second opinions to confirm the diagnosis and treatment plan.

Blood Cancer

Source(google.com.pk)
Blood Cancer Biography

Oncologist Siddhartha Mukherjee was treating one of his patients, a woman with advanced abdominal cancer who had relapsed multiple times, when she asked him what seemed like a simple question.

"She said, 'I'm willing to go on, but before I go on, I need to know what it is I'm battling,' " Mukherjee tells NPR's Terry Gross.

But, as Mukherjee explains, describing his patient's illness wasn't so simple. Defining cancer, he says, is something doctors and scientists have been struggling to do since the disease's first documented appearance thousands of years ago.

The Emperor of All Maladies: A Biography of Cancer
By Siddhartha Mukherjee
Hardcover, 592 pages
Scribner
List price: $30
"Cancer is not just a dividing cell," he says. "It's a complex disease: It invades, it metastasizes, it evades the immune system. So there are many, many other stages of [defining] cancer which are still in their infancy."

Mukherjee's new book, The Emperor of All Maladies: A Biography of Cancer, grew out of his desire to better understand the disease he treats, through examining the way cancer has been described and treated throughout history. He chronicles the ways therapies evolved, particularly in the 20th century, as more treatment options became available and scientists worked to understand the underlying genetic mutations that caused the disease.

Enlarge image
Siddhartha Mukherjee is a staff oncologist at Columbia University Medical Center.

Deborah Feingold via Scribner
"If there's a seminal discovery in oncology in the last 20 years, it's that idea that cancer genes are often mutated versions of normal genes," he says. "And the arrival of that moment really sent a chill down the spine of cancer biologists. Because here we were hoping that cancer would turn out to be some kind of exogenous event — a virus or something that could then be removed from our environment and our bodies and we could be rid of it — but [it turns out] that cancer genes are sitting inside of each and every one of our chromosomes, waiting to be corrupted or activated."

As the genetic understanding of cancer evolves, Mukherjee says, oncologists will be able to integrate that knowledge to develop more targeted treatment options — particularly as they find commonalities between different types of cancer.

"A breast cancer might turn out to have a close resemblance to a gastric cancer," he says. "And this kind of reorganization of cancer in terms of its internal genetic anatomy has really changed the way we treat and approach cancer in general."

Dr. Siddhartha Mukherjee is an assistant professor of medicine at Columbia University and a staff cancer oncologist at Columbia University Medical Center. His articles have been published in Nature, The New England Journal of Medicine, and The New York Times.

Excerpt: 'The Emperor Of All Maladies'
On the morning of May 19, 2004, Carla Reed, a thirty-year-old kindergarten teacher from Ipswich, Massachusetts, a mother of three young children, woke up in bed with a headache. "Not just any headache," she would recall later, "but a sort of numbness in my head. The kind of numbness that instantly tells you that something is terribly wrong."

Something had been terribly wrong for nearly a month. Late in April, Carla had discovered a few bruises on her back. They had suddenly appeared one morning, like strange stigmata, then grown and vanished over the next month, leaving large map-shaped marks on her back. Almost indiscernibly, her gums had begun to turn white. By early May, Carla, a vivacious, energetic woman accustomed to spending hours in the classroom chasing down five- and six-year-olds, could barely walk up a flight of stairs. Some mornings, exhausted and unable to stand up, she crawled down the hallways of her house on all fours to get from one room to another. She slept fitfully for twelve or fourteen hours a day, then woke up feeling so overwhelmingly tired that she needed to haul herself back to the couch again to sleep.

Carla and her husband saw a general physician and a nurse twice during those four weeks, but she returned each time with no tests and without a diagnosis. Ghostly pains appeared and disappeared in her bones. The doctor fumbled about for some explanation. perhaps it was a migraine, she suggested, and asked Carla to try some aspirin. The aspirin simply worsened the bleeding in Carla's white gums.

Outgoing, gregarious, and ebullient, Carla was more puzzled than worried about her waxing and waning illness. She had never been seriously ill in her life. The hospital was an abstract place for her; she had never met or consulted a medical specialist, let alone an oncologist. She imagined and concocted various causes to explain her symptoms — overwork, depression, dyspepsia, neuroses, insomnia. But in the end, something visceral arose inside her — a seventh sense — that told Carla something acute and catastrophic was brewing within her body.

On the afternoon of May 19, Carla dropped her two children with a neighbor and drove herself back to the clinic, demanding to have some blood tests. Her doctor ordered a routine test to check her blood counts. As the technician drew a tube of blood from her vein, he looked closely at the blood's color, obviously intrigued. Watery, pale, and dilute, the liquid that welled out of Carla's veins hardly resembled blood.

Carla waited the rest of the day without any news. At a fish market the next morning, she received a call.

"We need to draw some blood again," the nurse from the clinic said.

"When should I come?" Carla asked, planning her hectic day. She remembers looking up at the clock on the wall. A half-pound steak of salmon was warming in her shopping basket, threatening to spoil if she left it out too long.

In the end, commonplace particulars make up Carla's memories of illness: the clock, the car pool, the children, a tube of pale blood, a missed shower, the fish in the sun, the tightening tone of a voice on the phone. Carla cannot recall much of what the nurse said, only a general sense of urgency. "Come now," she thinks the nurse said. "Come now."

I heard about Carla's case at seven o'clock on the morning of May 21, on a train speeding between Kendall Square and Charles Street in Boston. The sentence that flickered on my beeper had the staccato and deadpan force of a true medical emergency: Carla Reed/New patient with leukemia/14th Floor/Please see as soon as you arrive. As the train shot out of a long, dark tunnel, the glass towers of the Massachusetts General Hospital suddenly loomed into view, and I could see the windows of the fourteenth floor rooms.

Carla, I guessed, was sitting in one of those rooms by herself, terrifyingly alone. Outside the room, a buzz of frantic activity had probably begun. Tubes of blood were shuttling between the ward and the laboratories on the second floor. Nurses were moving about with specimens, interns collecting data for morning reports, alarms beeping, pages being sent out. Somewhere in the depths of the hospital, a microscope was flickering on, with the cells in Carla's blood coming into focus under its lens.

I can feel relatively certain about all of this because the arrival of a patient with acute leukemia still sends a shiver down the hospital's spine — all the way from the cancer wards on its upper floors to the clinical laboratories buried deep in the basement. Leukemia is cancer of the white blood cells — cancer in one of its most explosive, violent incarnations. As one nurse on the wards often liked to remind her patients, with this disease "even a paper cut is an emergency."

For an oncologist in training, too, leukemia represents a special incarnation of cancer. Its pace, its acuity, its breathtaking, inexorable arc of growth forces rapid, often drastic decisions; it is terrifying to experience, terrifying to observe, and terrifying to treat. The body invaded by leukemia is pushed to its brittle physiological limit — every system, heart, lung, blood, working at the knife-edge of its performance. The nurses filled me in on the gaps in the story. Blood tests performed by Carla's doctor had revealed that her red cell count was critically low, less than a third of normal. Instead of normal white cells, her blood was packed with millions of large, malignant white cells — blasts, in the vocabulary of cancer. Her doctor, having finally stumbled upon the real diagnosis, had sent her to the Massachusetts General Hospital.

In the long, bare hall outside Carla's room, in the antiseptic gleam of the floor just mopped with diluted bleach, I ran through the list of tests that would be needed on her blood and mentally rehearsed the conversation I would have with her. There was, I noted ruefully, something rehearsed and robotic even about my sympathy. This was the tenth month of my "fellowship" in oncology — a two-year immersive medical program to train cancer specialists — and I felt as if I had gravitated to my lowest point. In those ten indescribably poignant and difficult months, dozens of patients in my care had died. I felt I was slowly becoming inured to the deaths and the desolation — vaccinated against the constant emotional brunt.

There were seven such cancer fellows at this hospital. On paper, we seemed like a formidable force: graduates of five medical schools and four teaching hospitals, sixty-six years of medical and scientific training, and twelve postgraduate degrees among us. But none of those years or degrees could possibly have prepared us for this training program. Medical school, internship, and residency had been physically and emotionally grueling, but the first months of the fellowship flicked away those memories as if all of that had been child's play, the kindergarten of medical training.

Cancer was an all-consuming presence in our lives. It invaded our imaginations; it occupied our memories; it infiltrated every conversation, every thought. And if we, as physicians, found ourselves immersed in cancer, then our patients found their lives virtually obliterated by the disease. In Aleksandr Solzhenitsyn's novel Cancer Ward, Pavel Nikolayevich Rusanov, a youthful Russian in his midforties, discovers that he has a tumor in his neck and is immediately whisked away into a cancer ward in some nameless hospital in the frigid north. The diagnosis of cancer—not the disease, but the mere stigma of its presence—becomes a death sentence for Rusanov. The illness strips him of his identity. It dresses him in a patient's smock (a tragicomically cruel costume, no less blighting than a prisoner's jumpsuit) and assumes absolute control of his actions. To be diagnosed with cancer, Rusanov discovers, is to enter a borderless medical gulag, a state even more invasive and paralyzing than the one that he has left behind. (Solzhenitsyn may have intended his absurdly totalitarian cancer hospital to parallel the absurdly totalitarian state outside it, yet when I once asked a woman with invasive cervical cancer about the parallel, she said sardonically, "Unfortunately, I did not need any metaphors to read the book. The cancer ward was my confining state, my prison.")

As a doctor learning to tend cancer patients, I had only a partial glimpse of this confinement. But even skirting its periphery, I could still feel its power—the dense, insistent gravitational tug that pulls everything and everyone into the orbit of cancer. A colleague, freshly out of his fellowship, pulled me aside on my first week to offer some advice. "It's called an immersive training program," he said, lowering his voice. "But by immersive, they really mean drowning. Don't let it work its way into everything you do. Have a life outside the hospital. You'll need it, or you'll get swallowed."

But it was impossible not to be swallowed. In the parking lot of the hospital, a chilly, concrete box lit by neon floodlights, I spent the end of every evening after rounds in stunned incoherence, the car radio crackling vacantly in the background, as I compulsively tried to reconstruct the events of the day. The stories of my patients consumed me, and the decisions that I made haunted me. Was it worthwhile continuing yet another round of chemotherapy on a sixty-six-year-old pharmacist with lung cancer who had failed all other drugs? Was is better to try a tested and potent combination of drugs on a twenty-six-year-old woman with Hodgkin's disease and risk losing her fertility, or to choose a more experimental combination that might spare it? Should a Spanish-speaking mother of three with colon cancer be enrolled in a new clinical trial when she can barely read the formal and inscrutable language of the consent forms?

Immersed in the day-to-day management of cancer, I could only see the lives and fates of my patients played out in color-saturated detail, like a television with the contrast turned too high. I could not pan back from the screen. I knew instinctively that these experiences were part of a much larger battle against cancer, but its contours lay far outside my reach. I had a novice's hunger for history, but also a novice's inability to envision it.