Wednesday, March 6, 2013

This is an official CDC HEALTH UPDATE
 
Distributed via the CDC Health Alert Network March 4, 2013, 16:30 ET (4:30 PM ET) CDCHAN-00342
 
Notice to Clinicians: Continued Vigilance Urged for
Fungal Infections among Patients Who Received
Contaminated Steroid Injections
  Summary
CDC continues to receive new reports of fungal infection among patients who were given injections of contaminated methylprednisolone acetate (MPA1) from the New England Compounding Center (NECC) in Framingham, Mass.   Most of these recent cases have been localized spinal or paraspinal infections (e.g., epidural abscesses) in patients, although new cases of meningitis or arachnoiditis also have been reported. Because many of these new cases are among patients with minimal symptoms, CDC is re-emphasizing the recommendation for clinicians to remain vigilant for fungal infections, especially in patients with mild or even baseline symptoms, and consider evaluation with magnetic resonance imaging (MRI) if clinically warranted. This Health Alert Network (HAN) notice provides the following:
·         Information about the current status of the outbreak;
·         Recommendations for clinical management and follow-up of exposed patients;
·         Information about new revisions to web-based interim clinical guidance (http://www.cdc.gov/hai/outbreaks/clinicians/guidance_cns.html); and
·         Notice of an upcoming CDC conference call to provide clinicians with additional diagnostic and treatment information.
 
Status of Fungal Disease Outbreak
As of March 4, 2013, a total of 720 cases, which includes 48 deaths, have been reported in 20 states. Current information about the outbreak, including case counts and distribution by state, and clinician and patient guidance, is available online at
http://www.cdc.gov/hai/outbreaks/meningitis.html.
Fungal meningitis, often with a mild clinical presentation, was the predominant clinical syndrome reported among case-patients during the first several weeks of the outbreak (figure). Over the past several months, there has been a marked decrease in reports of fungal meningitis, but CDC continues to receive reports of localized spinal and paraspinal infections, which include epidural abscess, phlegmon, arachnoiditis, and discitis.  Additionally, some of these newly identified case-patients had initially tested negative for signs of a fungal infection (either by lumbar puncture or MRI) and have subsequently developed fungal infection, indicating a prolonged incubation period.
 
After the recall of NECC steroid medications on September 26, state and local health departments identified almost 14,000 people in 23 states who were potentially exposed to the implicated MPA; of these, an estimated 11,000 individuals received spinal or paraspinal injections. Through active notification by clinics with assistance from states and CDC in early October, nearly all of these exposed persons were contacted at least once and informed of their risk for fungal infection as a result of receiving injections with contaminated medication.
Despite this and subsequent patient outreach efforts, CDC and public health partners remain concerned   about the potential for some exposed patients to have localized fungal infections that have gone unrecognized. These infections may be unrecognized because some patients have not continued to receive close clinical follow-up or because they have not recognized symptoms suggestive of a localized infection, which may be difficult to distinguish from their baseline chronic pain. 
As described in CDC’s HAN update on December 20 (http://emergency.cdc.gov/HAN/han00338.asp), MRI testing was done on 128 patients in Michigan, Tennessee, and North Carolina who had no previous evidence of infection and had new or worsening symptoms at or near the site of their spinal or paraspinal injection. Of these, 67 (52%) had findings suggestive of localized infection.  In addition, of 109 different patients reporting persistent but baseline symptoms at or near the site of their spinal or paraspinal injection, 15 (14%) also had abnormal MRI findings suggestive of infection, and 27 (25%) had non-specific enhancement of soft tissue or other paraspinal structures.  The clinical significance of these findings is unclear; however, there is a theoretical risk that failure to diagnose these infections in a timely fashion could result in poor outcomes for patients (e.g., neurologic compromise, osteomyelitis, or progression to meningitis
Patient and Clinician Recommendations
Early in the outbreak, CDC advised clinicians to closely monitor and evaluate patients who received injections of implicated MPA. Additional guidance was provided in HAN updates issued on November 20 (http://emergency.cdc.gov/HAN/han00335.asp) and December 20 (http://emergency.cdc.gov/HAN/han00338.asp). Because of the possibility that some patients may have unrecognized, localized fungal infections, CDC is re-emphasizing the following recommendations for patients who received a spinal or paraspinal injection with implicated MPA: 
 
Patients
Patients who received an injection in or near their spine from one of the three implicated lots of MPA1 and who have any symptoms at or near the site of their injection should seek evaluation by their medical provider for the possibility of a localized infection, such as an epidural abscess.  This includes patients who initially received steroid injections for pain and continue to have persistent baseline pain. 
 
Clinicians
As a part of continued monitoring of patients who received an injection with implicated MPA, clinicians should consider re-evaluating patients who received a spinal or paraspinal injection with implicated MPA for signs and symptoms suggestive of infection, including any symptoms at or near the site of their injection.  Because of the prolonged incubation period for these infections, this guidance pertains both to patients who have not been previously evaluated and to those who have already had a prior negative evaluation (e.g., normal cerebrospinal fluid profile, normal findings on MRI) but continue to have complaints:
 
-          In patients with new or worsening symptoms at or near the site of their injection, clinicians should obtain an MRI with contrast of the symptomatic area(s).
-          In patients with persistent but baseline symptoms, clinicians should consider obtaining an MRI with contrast of the symptomatic area(s) because the presentation of spinal or paraspinal infections can be subtle, and may be difficult to distinguish from a patient’s baseline chronic pain.
-          In some cases, radiologic evidence of abscess or phlegmon has become apparent on repeat MRI studies performed subsequent to an initially normal imaging procedure. Clinicians should therefore have a low threshold for repeat MRI studies in patients who continue to have symptoms localizing to the site of injection, even after a normal study. However, the optimal duration between MRI studies is unknown.
-          Clinicians should also consider reviewing MRI results with a neuroradiologist because of potential difficulties in interpreting imaging results for these patients.
 
Revised Clinical Guidance and Clinician Information Call
In response to input from expert consultants on fungal disease and physicians who have been treating patients affected by this outbreak, CDC has revised its Interim Treatment and Diagnostic Guidance for Central Nervous System and Parameningeal Infections Associated with Injection of Contaminated Steroid Products (http://www.cdc.gov/hai/outbreaks/clinicians/guidance_cns.html). The revisions include addition of new information on several topics, including:
 
-          Surgical management of parameningeal disease
-          Duration of antifungal treatment
-          Monitoring clinical status after cessation of antifungal treatment
-          Information on non-first-line medications (e.g., posaconazole or itraconazole)
 
A conference call for clinicians interested in obtaining additional information about the management and treatment of patients with fungal illness associated with this outbreak has been scheduled for March 13 at 5:00 p.m.  The presenter will be Tom Chiller, M.D., medical officer, CDC. Registration and call-in information and other details about the conference call will be available on CDC’s website http://www.cdc.gov/hai/outbreaks/clinicians/index.html.
­___________________________________________________________
1NECC lots of methylprednisolone acetate (PF) 80mg/ml:
Methylprednisolone Acetate (PF) 80 mg/ml Injection, Lot #05212012@68, BUD 11/17/2012
Methylprednisolone Acetate (PF) 80 mg/ml Injection, Lot #06292012@26, BUD 12/26/2012
Methylprednisolone Acetate (PF) 80 mg/ml Injection, Lot #08102012@51, BUD 2/6/2013
 
The Centers for Disease Control and Prevention (CDC) protects people's health and safety by preventing and controlling diseases and injuries; enhances health decisions by providing credible information on critical health issues; and promotes healthy living through strong partnerships with local, national, and international organizations.
____________________________________________________________________________________
 
Categories of Health Alert Network messages:
Health Alert         Requires immediate action or attention; highest level of importance
Health Advisory   May not require immediate action; provides important information for a specific incident or situation
Health Update     Unlikely to require immediate action; provides updated information regarding an incident or situation
HAN Info Service                Does not require immediate action; provides general public health information

##This message was distributed to state and local health officers, public information officers, epidemiologists, HAN coordinators, and clinician organizations##
 
SUPPRESSION OF NF-kB IS ONE OF THE DOMINANT EVENT IN ACUTE MYELOID LEUKEMIA,

As we explore the major genes involved in Acute Myeloid leukemia, we quickly realize that the main initiating event is located in the Core binding factors or complex proteins located at the Histone-DNA.  At this level we already uncovered that the nature of molecule interacting and  involved are considerably important, and fundamentally different when you speak about Hematological neoplasm versus solid tumor.
Globally, they appear to be several levels of action:

1.  Nature of components of Histone (H1A, H2A etc..) as cover of DNA. 
2.  Complex involved in Histone remodeling  (H1Ax)
3.  Portion Alpha-subunit of protein complexes attached to the DNA to control its expression, and here we find the RUNX which control Hematologic differentiation
4. Portion of Alpha subunit that actually ensure just clear attachment to DNA so that the Histones do not run in the nuclear  solution.  But be careful in fact most of the time if not always, the place of attachment of histone is not random and varies according to nature of tissue involved.  That is Histones attachment contribute to gene silencing and tissue differentiation.
5. Then there Beta subunits which send tentacles dealing with
    5.1-pure Histone Deacyl transferase activity
    5-2- DNA uncoiling and coiling
    5.3- Interaction with Regulators
    5.4-Interactions with cytoplasmic  signal trasductions (MAPK, FOS/c-JUNK, RAS,PI1K ,VEGF)
    5.5-output back to the cytoplasm to inhibit or activate regulators of signal transduction.  The control of signal transduction pathways is done through enzymes production but through activation of switches (E3, SOS) and through control of Ubiquitination and the MDM2
   5.6- DNA replication controlled through P53, and check-point control molecules.
    5.7-signal to Mitochondria, the Ribosome AND AT C-MYC
  etc. (Transcription factors)
The Centrosome have a DNA attachment portion and an endonucleases portion and some endonucleases find their way to  this area of histone-DNA.

Suffice is to say that in AML, the RUNX is involved to ensure Hematologic differentiation, Many regulators are involved, but suppression of the NF-kB and therefore suppression of the TGF is an significant find!
One now speculate as to why it is so.  AND WITHOUT HESITATION, ONE CONCLUDE THAT IT IS BECAUSE AML DOES NOT NEED TO FORM A MASS!  THE SUPPRESSION OF TGF CAN BE INSUFFICIENT HOWEVER, AND A GRANULOCYTIC SARCOMA IS CREATED BUT THIS IS RARE.

(CLEARLY SOME OF THE CONCEPTS PRESENTED HAVE STILL TO BE FULLY VISITED BY RESEARCHER AND READERS-READ MORE TO ESTABLISH THIS IS SO!)

IT IS INTERESTING TO NOTE THAT WHILE IN THE STRUGGLE IS AT THE RUNX IN AML, IN BLADDER CANCER THE TGF IS IS FULL SWING, DNA REPAIR IS IN FULL SWING, BUT ALSO EVENTS AT THE HISTONE MODULATION AS ALSO IN FULL SWING

Tuesday, March 5, 2013

Bladder cancer in 2012:
Prevalence 73.5 Thousand patients in the USA
Deaths 14.9 thousands died with this disease.
Only 15% of patients have localized disease
50% of patients who have muscle invasion will eventually progress, Median survival in Metastatic disease is 14 months.
Smoking is an established risk factor for Bladder cancer

When the Bladder Muscle is invaded, Giving 3 cycles of chemotherapy (MVAC) before surgery is standard of care and lead to a 77 months median survival Versus 46 months with surgery alone (SWOG 8710.) . But only 20% of people receive Neoadjuvant Chemotherapy.   Mostly because the disease affect the elderly who's perfomance is judged poor for the chemotherapy, because of renal failure which preclude use of Cisplatin, and related comorbidilties which would make chemotherapy hazardous.

Genes reported in Bladder cancer:

MRE11
RAD50
NBS1
ATM
_______________________________________________-----These 4 genes all involves a close pathway.   Under chemical effect there is breakage of double chains DNA in bladder cells  The DNA Break trigger stimulate ATM by phosphorylation or similar activation, MRE1-RAD50-NBS1 is a complex molecule which must allow repair of the breakage by exposing it through "histone remodeling".  Mutations here will hamper this sequence of Activity.  "Breakage of DNA" and HOP, P53 activation is not far.  This is why intact P53 or wild type marks an early cancer. and Mutation of P53 is more a sign of an advanced Bladder cancer.
-----------------------------------------------------------------------------------------
H2AX-during Histone remodeling H2Ax replace H2A and participate in the triggering of check point arrest and therefore a key point to the Histone complex activity which is to control DNA transcription.
------------------------------------------------------------------------------------------------------
CSS (Calpain family) contain DeK1 and CysPc and other anchors/molecules (Now you are going closer to treating AML)
Resistance to Cisplatin: ERCC1
Bcl-2
Overexpression of HRAS (in early disease)
70% have Mutation in FGFR3
P53 and Rb more advanced disease
P21, and P16 are also bad prognosis predictors.

NMP22 may be identified in the Urine for early detection of superficial bladder cancer



A GOOD READING ABOUT SARCOMA

Volume 2011 (2011), Article ID 483154, 13 pages
doi:10.1155/2011/483154
Review ArticleLiposarcoma: Molecular Genetics and Therapeutics

Rachel Conyers, Sophie Young, and David M. ThomasSarcoma Genomics & Genetics, Peter MacCallum Cancer Centre, 12 St Andrews Place, East Melbourne, VIC 3002,  AustraliaReceived 16 September 2010; Accepted 29 October 2010Academic Editor: Stephen Lessnick Copyright © 2011 Rachel Conyers et al.

This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Sarcomas are a group of heterogeneous tumours with varying genetic basis. Cytogenetic abnormalities range from distinct genomic rearrangements such as pathognomonic translocation events and common chromosomal amplification or loss, to more complex rearrangements involving multiple chromosomes. The different subtypes of liposarcoma are spread across this spectrum and constitute an interesting tumour type for molecular review. This paper will outline molecular pathogenesis of the three main subtypes of liposarcoma: well-differentiated/dedifferentiated, myxoid/round cell, and pleomorphic liposarcoma. Both the molecular basis and future avenues for therapeutic intervention will be discussed.1. IntroductionAn estimated 13,000 people were diagnosed with soft tissue and bone sarcoma in 2009 in America, of which liposarcomas constitute 20% [1, 2]. Despite their rarity these tumours have substantial morbidity and mortality, depending on histological subtype, tumour location, and volume with retroperitoneal sarcomas having particularly poor prognosis [3–9]. Liposarcomas may be classified morphologically into 3 main subtypes consisting of:  well-differentiated liposarcoma/de-differentiated liposarcoma (WD/DDLPS), myxoid/round cell liposarcoma (MLPS) and pleomorphic liposarcoma (PLPS) [10]. The morphological diversity of liposarcoma reflects the great variation in biological behaviour ranging from tumours with low metastatic potential, that is, WDLPS, to tumours with high propensity to metastasise, that is, the round cell (RC) variant of MLPS or PLPS [11]. In addition to histological characteristics, anatomical location impacts upon prognosis, given that local control is a prime concern for curative intent. Treatment is multimodal with surgical removal and radiotherapy used as cornerstones for local control, along with chemotherapy for systemic disease. Few therapeutic options are available for aggressive local or metastatic disease. Chemotherapy sensitivity varies considerably between subtypes with higher response rates in MLPS compared with WD/DDLPS (48% versus 11%) [12]. MLPS tumours are also highly radiosensitive [13, 14]. Given the small subgroup that is chemo-sensitive, and the overriding lack of chemo-curative disease there are avenues and a need for novel molecular therapies. A recent histological and molecular review of 163 liposarcoma and lipomas at the Netherlands Cancer Institute resulted in 23% of tumours being reclassified based on cytogenetic information. This highlights the importance of molecular classification in these tumours and genetic alterations now considered an integral part of the WHO classification [15]. It is hoped that further insight into the molecular characteristics of liposarcomas will allow for accurate subclassification, whilst providing a platform for molecular therapies to be included in the current treatment approach. This paper will outline the current molecular basis of liposarcoma and potential strategies for therapeutic intervention.2. Well- and De-differentiated LiposarcomaWDLPS represents 40%–45% of all diagnosis of liposarcoma [16]. It is classified as a low-grade neoplasm; it is rarely metastatic and has a low recurrence rate (10%) occurring most often in the retroperitoneum and limbs. The World Health Organization (WHO) classifies WDLPS into three main subtypes: adipocytic, sclerosing, and inflammatory. Adipocytic (lipoma-like) liposarcoma is composed of mature adipocytes, which exhibit variation in cell size and focal nuclear atypia and hyperchromasia [16]. The sclerosing subtype shows scattered distinctive bizarre stromal cells associated with rare multivacuolated lipoblasts set in a fibrillary collagenous background [16]. Finally, the inflammatory subtype shows polyphenotypic lymphoplasmacytic infiltrate, with a B-cell predominance. Less is known about this rare subtype [16–18]. DDLPS represents progression from low grade to high-grade nonlipogenic morphology within a WDLPS. DDLPS is more aggressive and exhibits an increased rapidity of disease in contrast to WDLPS, with a metastatic rate of 10%–20% and overall mortality of 50%–75% [4, 7, 19]. In respect to tumour site, retroperitoneal tumours appear to have a worse prognosis [19]. Histologically, DDLPS consists of a WDPLS with a nonlipogenic component, either high-grade, most often resembling malignant fibrous histiocytoma (MFH), or low-grade resembling fibromatosis or low-grade myxofibrosarcoma. The presence of transition from WDLPS to DDLPS is used to differentiate between DDLPS and these other lesions [4, 7, 11, 19–21].2.1. Molecular GeneticsA characteristic feature of WD/DDLPS is the presence of supernumerary ring and/or giant rod chromosomes [22]. These chromosomes contain amplified segments from the 12q13–15 region that can be identified with fluorescence in situ hybridization (FISH) and comparative genomic hybridization (CGH) [23]. Intensive research has identified several oncogenes residing in this region including MDM2, CDK4, HMGA2, TSPAN31, OS1, OS9, CHOP and GLI1 [11, 23–25]. The most compelling evidence to date demonstrates an oncogenic role in WD/DDLPS for MDM2, CDK4, HMGA2 and TSPAN31. Additional amplification events may also play a role in liposarcoma genesis, for example, c-Jun in the de-differentiation process [26].MDM2 amplification is a key feature of WD/DDLPS and is amplified and overexpressed in a number of other cancers, highlighting its importance in tumorigenesis (as reviewed [27]). MDM2 encodes a negative regulator of the tumour suppressor, p53. MDM2 binds to the transcription activation domain of p53, within an N-terminal hydrophobic pocket [28], blocking p53-dependent transcription [29–33] and recruitment of transcription coactivators [28]. MDM2 also acts as a ubiquitin ligase targeting p53 for proteasomal degradation through both cytoplasmic and nuclear proteasomes [34–36]. MDM2 is involved in its own auto-degradation to prevent MDM2 activity inhibiting p53 during times of cellular stress [37]. Thus MDM2 maintains tight control on cellular p53 levels through multiple mechanisms (see Figure 1) [38, 39]. Therapeutically this is important, as MDM2 inhibitors aim to reactivate p53 and thus allow it to actively induce cell death in response to appropriate stressors [40]. In addition to a functional downstream p53 signalling pathway, MDM2 amplification is a predictor of sensitivity to current MDM2 antagonists [40]. Amplification of MDM2 and mutation of p53 appear to be mutually exclusive events in WDLPS, but have been reported in DDLPS [41, 42]. p53 mutations have been associated with the de-differentiation process from WDLPS to DDLPS [41].  Pilotti et al. reported upon a subgroup of WD/DDLPS tumours. Retroperitoneal WD/DDLPS demonstrate mutual exclusivity between MDM2 amplification and p53 mutation. In non-retroperitoneal DDLPS, p53 mutations occur in the absence of MDM2 amplification suggesting involvement in the de-differentiation process [41].483154.fig.001Figure 1: MDM2 binds to the transcriptional activation domain of p53, blocking transcription. MDM2 functions as a ubiquitin ligase, facilitating proteasomal degradation of p53. MDM2 releases p53 in response to cellular stress and p53 translocates to the nucleus where it acts as a transcription factor to enable growth arrest and apoptosis.MDM2 is the most frequent amplification in WD/DDLPS (close to 100%) however CDK4 is shown to be amplified in over 90% of cases [16, 43, 44]. Given its role in the cell cycle and the frequency of amplification, CDK4 has been well researched in WD/DDLPS. The CDK4 gene encodes a 33-kD protein that forms complexes with the cyclin D family, to enable G1-S transition [45]. These CDK4/Cyclin D complexes phosphorylate pRb (encoded by RB1), with resultant activation of E2F target genes including E-type cyclins (see Figure 2) [46–48]. It has been suggested that CDK4 provides a selection advantage in WD/DDLPS and may contribute to transformation as CDK4 negative WDLPS exhibit more favorable prognostic features [46]. Coamplification of MDM2 and CDK4 is a common feature of WD/DDLPS and may result in proliferation through combined effects upon p53 and the cell cycle [49, 50]. Interestingly, the rearrangements of chromosome 12 on the giant rod chromosome are discontinuous and MDM2 and CDK4 may belong to different amplicons [51, 52]. Several studies [43, 53, 54] have suggested that immunohistochemical staining for both CDK4 and MDM2 may provide a useful diagnostic marker, although FISH and quantitative polymerase chain reaction (qPCR) are more effective. Although MDM2 and CDK4 are useful markers to aid in diagnosis, overexpression of these markers is not unique to WD/DDLPS [43, 54]. Further, the amplification and over-expression of CDK4 and MDM2 does not distinguish WDLPS from DDLPS [16, 23, 41]. 483154.fig.002Figure 2: Cyclin dependent kinase CDK4 binds with cyclin D to form active complexes. This results in phosphorylation of Rb and dissociates pRb from the pRb-E2F complex. E2F binds DNA to upregulate transcription of genes required to progress to S phase.HMGA2 is similarly located on 12q and frequently amplified in WD/DDLPS. This is a member of the high-mobility group of proteins [55, 56]. Previously referred to as HMGIC, it encodes an architectural transcription factor capable of remodeling DNA [57–59]. A direct role for HMGA2 in cellular transformation is demonstrated by NIH3T3 neoplastic transformation with the overexpression of HMGA2 [60]. In human sarcomas during chromosomal rearrangement, HMGA2 is fused to distant sequences, commonly occurring on other chromosomes and loses its 3′ translated end that also contains sites for Let-7 microRNAS [57]. Further support for HMGA2 involvement in adipogenic neoplasm development includes the xenograft model by Arlotta et al. [55] that showed mice expressing C-terminal truncated HMGA2 developed lipomas. Interestingly HMGA2 is frequently coamplified with MDM2 in human malignant tumours [57, 61], particularly WDLPS and DDLPS [52]. This raises the possibility that HMGA2 and MDM2 have a cooperating role in WD/DDLPS. Also included within the chromosome 12 q13–15 region is the transmembrane superfamily gene, sarcoma amplified sequence (SAS or TSPAN31) gene [62, 63]. TSPAN31 was originally identified and cloned from an amplified sequence in a malignant fibrous histiocytoma [63]. It has been identified in other subtypes of sarcoma, particularly de-differentiated liposarcoma [64, 65], although its precise role in the de-differentiation process is not well delineated. Forus et al. [66] showed TSPAN31 was as frequently amplified as MDM2 in 98 sarcomas. Both TSPAN31 and MDM2 were amplified in 8 of 11 liposarcoma samples, with MDM2 amplified alone in one additional tumour. WDLPS and DDLPS have shown co-amplification of 1q21-q22 and/or 12q21-q22 [11, 16, 23], along with amplification of chromosome 1(1q21-q23). Chromosome 1 amplified sequences include COAS1, COAS2 and COAS3 [67]. Nilsson et al. showed co-amplification of both COAS and MDM2 in 12/18 lipomatous tumours [68].  The biological function of the COAS genes remains a subject for study.Recent studies into the WDLPS de-differentiation process have suggested a role for the c-Jun N-terminal kinase (JNK) pathway. Co-amplification of 1p32 and 6q23, that contain c-Jun, and Apoptosis Signaling Kinase 1 (ASK1), are seen in DDLPS but not WDLPS [69]. The proto-oncogene c-Jun encodes part of the activator protein transcription factor (AP-1) complex involved in cell proliferation, transformation and apoptosis [70]. ASK1 activates JNK [71, 72] ultimately leading to c-Jun activation and PPARγ inactivation. PPARγ is involved in the adipocytic differentiation process and its inhibition may result in de-differentiation. A further role for c-Jun in the de-differentiation process is demonstrated by overexpression in a 3T3-L1 adipocytic tumour xenograft model. Transfection of c-Jun into 3T3-L1 cells in vitro delays adipocytic differentiation [26].3. Myxoid LiposarcomaMLPS is the second most common subtype of liposarcoma and accounts for more than one third of liposarcomas and 10% of all adult soft tissue sarcomas. MLPS is characterized by the presence of spindle or ovoid cells set in a myxoid stroma with signet ring lipoblasts and a distinctive chicken-wire pattern vasculature. The presence of areas with greater cellularity, known as round cell (RC) de-differentiation, is associated with a worse prognosis [73]. Unusual sites of metastasis are common in MLPS with a propensity to metastasize to soft tissue and bone rather than lung [74, 75]. Thirty-one percent of MLPS patients develop metastasis with bone metastases constituting 56% of these [74]. MLPS exhibits inferior survival compared to other low-grade sarcoma subtypes with a 5-year disease survival rate of 85% [76, 77]. MLPS without RC is particularly radiosensitive with good local control rates with patients treated with adjuvant or neoadjuvant radiotherapy approaching 98% 5-year local control [13, 78].3.1. Molecular GeneticsMLPS is characterised by the recurrent translocation t(12;16)(q13;p11) that results in the FUS-CHOP gene fusion that is present in over 95% of cases [79, 80]. In most cases, the amino terminal domain of FUS (also known as TLS) is fused to C/EBP homologous protein (CHOP, also known as DDIT3 or GADD153). In rare cases, an alternative translocation event is found t(12;22)(q13;q12) that results in formation of the novel fusion oncogene where EWS takes the place of FUS [81, 82]. There is strong evidence for these translocations to be the primary oncogenic event in MLPS as these tumours have a relatively normal karyotype, the exception being a few recurrent cases of trisomy 8 [83]. In addition, several growth factor pathways have been implicated in MLPS pathogenesis [84–86].There are currently 11 different FUS-CHOP chimeras and 4 different known EWS-CHOP fusion genes. In the most common variants, a portion of the amino terminus of FUS is fused to the entire coding region of CHOP. The FUS-CHOP transcript type does not appear to have a significant impact upon clinical outcome, and RC content, necrosis and p53 expression remain stronger predictors of clinical outcome [79, 87]. There is evidence that the fusion transcript type may influence response to therapy although the studies are hindered by sample size [88–90]. Understanding how the FUS-CHOP fusion causes MLPS and uncovering any further molecular abnormalities in the disease will aid in development of novel targeted therapies.FUS belongs to the FET family of RNA-binding proteins that consists of FUS, EWS, and TAF15 as well as the closely homologous, Drosophila SARFH (Cab) [80, 91, 92]. These structurally and functionally related RNA-binding proteins are composed of an SYGQ-rich amino terminus, an RNA recognition motif, a zinc finger motif, and at least one RGG rich repeat region [93, 94]. FET proteins are expressed in most human tissues and appear to be regulated following differentiation in neuroblastoma cells and spontaneously differentiating human embryonic stem cells [95].Both FUS and EWS have been shown to localize to the nucleus and the cytoplasm, bind RNA, and are also involved in nucleo-cytoplasmic shuttling [96–98].  The FET family associate with various complexes involved in the induction of transcription, including RNA polymerase II (RNAPII), which regulates transcription and TFIID complexes, that binds DNA as part of the transcriptional machinery [91], implicating both FUS and EWS in transcriptional control. In addition, FUS has recently been shown to repress transcription of RNA polymerase III (RNAPIII), suggesting a broader role in regulation through multiple different mechanisms [99]. Noncoding RNAs are capable of allosterically modifying FUS in response to DNA damage to inhibit the transcription factor CREB-binding protein (CBP) and p300 histone acetyltransferase activity, resulting in transcriptional inhibition at the cyclin D1 promoter in cell lines and shows a further role for FUS in transcriptional control [100].  FUS has also been implicated in the DNA damage response as a downstream target of ATM, which can detect and coordinate DNA repair [101].CHOP is induced in response to endoplasmic reticular stress and is involved in mediating cell death in response to such stress stimuli [102]. CHOP also plays a role in regulating differentiation in adipocytes by interfering with the process in response to metabolic stress [103]. Adipocytic differentiation is dependent on the coordinated expression of a group of transcription factors, the CCAAT/enhancer-binding protein (C/EBP) family of proteins [104]. The C/EBP family consists of six members from C/EBPα to ζ, and they require dimerisation to bind DNA and can form homodimers or heterodimers. CHOP is capable of binding to the C/EBP family members through their highly conserved leucine zipper domain and inhibiting their function. The leucine zipper dimerization domain and the adjacent basic region in CHOP are required for NIH-3T3 transformation with FUS-CHOP, highlighting the requirement for functional DNA binding and dimerization for FUS-CHOP induced oncogenesis [105]. As C/EBPα and C/EBPβ play an important role in the adipogenic differentiation and are regulated by CHOP, it is possible FUS-CHOP may interfere in cellular differentiation. In support, various studies suggest that FUS-CHOP functions by inhibiting adipogenesis and maintaining immature adipocytes in a continuous cycle of proliferation without differentiation [106–108]. Introduction of FUS-CHOP into mice, where expression of the transgene is driven by the ubiquitously expressed elongation factor 1α (EF1α) promoter, results specifically in liposarcomas with inherent induction of adipocyte specific genes such as PPARγ [109].  Further evidence of adipogenic differentiation block resulting from FUS-CHOP expression was shown in vitro where mice expressing FUS-CHOP under the control of the aP2 promoter, which is a downstream target of PPARγ expressed in immature adipocytes, failed to develop liposarcomas, indicating interference between PPARγ and aP2 activation [107].An emerging clinically relevant targetable pathway in MLPS involves the receptor tyrosine kinases (RTKs) MET, RET, and the PI3K signaling cascade (see Figure 3).  RET is overexpressed in MLPS compared to normal fat [84] and high expression has been correlated with poor metastasis free survival in MLPS [108].  RET, IGF1R and IGF2 are highly expressed in MLPS and promote cell survival through both the PI3K/Akt and Ras-Raf-ERK/MAPK pathways [85, 86]. A panel of tyrosine kinases including PDGFRB, EGFR, MET, RET, and VEGFR2 are activated in both treated (with chemotherapy/radiotherapy or Trabectedin) and untreated cases of human MLPS [110]. In addition to activation of MET in clinical MLPS specimens, MET and the ligand HGF are potentially regulated by FUS-CHOP. Both MET and HGF are highly expressed in mesenchymal progenitor cells transfected with FUS-CHOP in a disease mimicking allograft mouse model [111]. In a small clinical cohort, specific Akt phosphorylation was observed in the RC variant and 2 treated cases that harboured PTEN mutations, implicating RTK pathways signaling through Akt in MLPS [110].  FLT1 (that encodes the VEGFR1 protein) is expressed as an indirect downstream effect of FUS-CHOP expression in both FUS-CHOP transfected HT1080 (fibrosarcoma) and MLPS cell lines however, VEGFR tyrosine kinase inhibitors did not have a notable impact on proliferation in MLPS cell lines indicating a separate role in these cells [112, 113]. 483154.fig.003Figure 3: The PI3K pathway is highly active in MLPS, and this is potentiated at least in part by overexpression, and/or activation through RTKs such as MET, RET and VEGFRs. Upon ligand binding, RTKs activate downstream activation of genes involved in multiple cell processes such as cell survival, proliferation, and angiogenesis. These signals are mediated through the PI3K/Akt pathway and also through RAS. PIK3CA and PTEN mutations and Akt activation have also been documented in MLPS.Akt activation, particularly in the RC variant, suggests a role for phosphoinositide 3-kinases (PI3K) [110]. PI3Ks are activated upon phosphorylation of membrane bound receptor tyrosine kinases. PI3K can activate many proteins including the protein serine-threonine kinase Akt, which when phosphorylated causes downstream activation and ultimately cell growth, cell cycle entry, and subsequently survival. The PI3K holoenzyme complex is composed of both a catalytic and regulatory subunit. The catalytic subunit, PIK3CA, encodes the p110α isoform and is commonly mutated in various cancer types including breast, colon, brain and gastric malignancies [114, 115]. A recent study showed 18% of MLPS patients (
   
       
           

       
   
) had PIK3CA mutations in either the helical (E542K and E545K) or kinase (H1047L and H1047R) domain. The presence of a PIK3CA mutation was associated with a shortened disease specific survival [116]. Barretina et al. also showed one tumour with a homozygous PTEN mutation. PTEN is a tumour suppressor that dephosphorylates phosphoinositide substrates to negatively regulate the Akt signaling pathway [117], demonstrating more mechanisms for perturbation of the pathway.4. Pleomorphic LiposarcomaPLPS accounts for only 5% of liposarcomas and occurs mainly within the 55–65 year-old group [8, 118, 119]. PLPS mortality is 40% with no current clinical or pathological predictors of outcome [8, 120]. Histologically PLPS are similar to MFH with the addition of lipoblasts. Histology reveals a disorderly growth pattern, extreme cellularity, and cellular pleomorphism including bizarre giant cells [121]. Lesional cells are polygonal with pale eosinophilic cytoplasm and poorly demarcated boundaries. These lesional cells are interspersed with giant lipoblasts containing enlarged hyperchromatic, angular or globular nuclei [121, 122].4.1. Molecular GeneticsMolecular studies of PLPS are limited by the scarcity of this disease. Tumours tend to show complex arrangements including gains: 1p, 1q21-q32,2q, 3p, 3q, 5p12-p15, 5q, 6p21, 7p, 7q22 (see reviews) [118, 123, 124]. reported literature shows losses i of 1q, 2q, 3p, 4q, 10q, 11q, 12p13, 13q14, 13q21-qter, 13q23-24, (see reviews) [123–125], Taylor et al. described that 60% of PLPS have a deletion of 13q14.2-q14.3, a region that includes the tumour suppressor RB1 [123]. Also amplified in PLPS, the mitotic arrest deficient (MAD2) may also play a critical role [126, 127]. As reported by Singer et al. [126], MAD2 was found to be over-expressed 13 fold in comparison to normal fat, although small sample size (
   
       
           

       
   
) must be appreciated. As reported by Taylor et al. [123] additional deletions in PLPS include 17p13 and 17q11.2, where p53 and the sarcoma associated tumour suppressor gene, neurofibromatosis type 1 (NF-1) are located. Consistent with these observations, Barretina et al. [116] showed 16.7% of PLPS cases had mutations identified in p53, which are rarely seen in MLPS and WD/DDLPS.5. Therapeutic Implications in LiposarcomaThe current modalities available  (chemotherapy, surgery and radiotherapy) for the treatment of liposarcoma are limited, creating a need to identify novel therapeutics.5.1. MDM2 AntagonistsGiven MDM2 is consistently amplified in WD/DDLPS, and sensitivity to MDM2 antagonists (such as Nutlin-3a) is predicted by MDM2 amplification and an intact wild-type p53, it is an appealing therapeutic target [40]. First generation MDM2 inhibitors work via blocking the p53/MDM2 interaction. Nutlin-3a was heralded as one of the most promising MDM2 antagonists when it was shown to activate wild type p53 and induce cell cycle arrest and apoptosis in cancer cell lines [40]. These cell lines included osteosarcoma with amplified MDM2 [40, 128]. Nutlins require wild-type p53 and a functional downstream p53 pathway to be effective [128]. Müller et al. [40]  showed downstream p53 dependent transcription and apoptosis in liposarcoma cell lines treated with Nutlin-3a [40]. Translation from in vitro to attractive in vivo therapeutic intervention requires that drugs pass Phase I requirements. Shangary et al. [129] designed spiro-oxindoles as a new class of inhibitors of the MDM2-p53 complex.  Spiro-oxindoles bind to MDM2 with high affinity and activates the p53 pathway, inhibiting the growth of neoplastic cell lines with wild-type p53 [129, 130]. MI-219, the lead compound in this class, demonstrates greater potency along with a superior pharmacokinetic profile than Nutlin-3a [129, 131]. MI-219 has been shown to stimulate rapid p53 activation in tumour xenograft tissues with resultant inhibition of cell proliferation [131]. Studies using both Nutlin-3a and MI-219 show a p53 and p21 dependent cell cycle arrest in normal cells, along with p53 dependent cell death specifically in tumour cells [128, 129, 131, 132]. The ability of Nutlin-3a to induce apoptosis in tumours is variable, and osteosarcoma cell lines lacking MDM2 amplification are resistant to apoptosis [131]. Importantly, Nutlin-3a and MI-219 do not cause visible toxicity to animals, as assessed at necropsy [128, 129, 133]. Two oral MDM2 inhibitors have recently entered the clinical setting [134], JNJ-26854165 (Ortho Biotech; Johnson & Johnson) [135] and R7112 (Hoffmann-La Roche) [136]. Both agents are available in advanced stage or refractory solid tumours Phase I trials [134]. In addition, AT-219 (a derivative of MI-219) is in preclinical studies with phase I trials planned [134]. Of relevant interest, an MDM2 antagonist RO5045337 is about to recruit for a Phase I trial in liposarcoma patients [137].5.2. CDK4 AntagonistsTargeting CDK4 is an attractive therapeutic strategy given its frequent overexpression in WD/DDLPS [138]. A number of CDK4 inhibitors are in the early pre-clinical development or Phase I and II trials [139]. First generation pan-CDK inhibitors include Flavopiridol and Seleciclib (R-Roscovitine), inhibiting CDK1, CDK2, CDK4, CDK6, CDK7, and CDK1, CDK2, CDK7 and CDK9 respectively [140].  Flavopiridol causes arrest in G1 and G2 phases in a range of solid tumour cell lines [139, 141, 142]. Flavopiridol is more potent if tumour cells are in S phase. Matranga and Shapiro [143] demonstrated recruitment to S phase using hydroxyurea, gemcitabine and cisplatin, followed by flavopiridol resulting in sequence-dependent cytotoxic synergy [143–145]. Flavopiridol and Seliciclib have been investigated in Phase I/II trials for haematological and solid tumours including sarcomas. Trials include Flavopiridol as a single agent and in combination with taxanes where synergism has been noted [141]. Both Flavopiridol and Seleciclib have shown disappointing results relating to clinical outcome and intolerable side effects [146, 147]. Newer generation CDK inhibitors include PD0332991, P27600, ZK 304709, R 547 and P1446A05.  All are available in Phase I and II solid tumour trials [146]. PD0332991 is one of two more selective CDK inhibitors specific for CDK4 and CDK6. Preclinical data showed inhibition of cell growth through G1 arrest in pRb-positive tumour cell lines and antitumorigenic effects in xenograft models of colon carcinoma [148]. PD0332991 is available in Phase I and Phase II trials for solid and haematological malignancy. Finally, P1446A05 is the only single CDK4 selective inhibitor available [146]. No pre-clinical data is publicly available for this compound; however, it has been released as a Phase I drug for refractory solid tumour and haematological malignancies [146].5.3. PPARγ Ligand AgonistsA critical regulator of terminal differentiation for the adipocytic lineage is a nuclear receptor peroxisome proliferator-activated receptor γ [149–151]. PPARγ is an attractive target in undifferentiated lipomatous tumours such as DDLPS and MLPS.  PPARγ forms a heterodimeric complex with the retinoid X receptor (RXR).  This complex regulates transcription of adipocyte-specific genes by binding sites on DNA. Agonist ligands for the PPARγ receptor have been shown to induce terminal differentiation of normal preadipocytes in human liposarcoma cells in vitro [149].    A Dana-Farber Cancer Institute Phase II clinical trial used Troglitazone, a synthetic PPARγ ligand, in patients with high-grade liposarcoma. This trial enrolled three patients. All patients showed histologic and biochemical differentiation in vivo, with reduction in immunohistochemical expression of proliferation marker Ki-67 [149]. A more recent study with 12 patients with Rosiglitazone, belonging to the same class of drugs (thiazolidinediones) as Troglitazone, was not as promising, with median progression free survival of 5.5 months. Treatment did not produce any convincing adipocytic differentiation with no correlation between the high expression of differentiation genes that was found in two patients, and clinical response [152].5.4. Trabectedin (ET-743)Trabectedin (also known as Ecteinascidin or ET-743) is an antitumor drug isolated from the Caribbean marine tunicate, Ecteinascidia turbinata [153]. Trabectedin is an approved second-line agent for advanced soft tissue sarcoma and has been shown to be exquisitely sensitive to Trabectedin in Phase II clinical trials [154, 155]. The drug is a tetrahydroisoquinoline alkaloid whose main mechanism of action is through binding to the DNA minor groove with promoter and sequence specificity; however, it has also been shown to have effects on promoters that are regulated by major groove binding transcription factors [156–158]. Trabectedin does not appear to effect transcription of FUS-CHOP, but has been shown to dissociate the aberrant transcription factor from promoters of its target genes resulting in removal of the differentiation block by activating a differentiation cascade through the C/EBPs [88]. Trabectedin relies on intact nucleotide excision repair (NER) machinery and induces lethal DNA strand breaks in a transcription-couple NER dependant manner [159–161]. It has been suggested that these breaks are repaired by homologous recombination (HR), as HR-deficient cells, such as BRCA2 mutants, are 100 fold more sensitive to Trabectedin [162]. This effect is specific to HR- mediated double strand break repair as defects in the alternative pathway using nonhomologous end joining do not result in the same degree of Trabectedin sensitivity [161, 162]. FUS-CHOP modulates immune genes by activating NF-κB controlled cytokines IL-6 and IL-8 in a C/EBPβ- dependent manner [163, 164]. Proinflammatory cytokines and growth factors such as CCL2, CXCL8, IL-6, VEGF and PTX3 are highly expressed in both xenograft MLPS models and patient tumours. Trabectedin has been shown to reduce expression and production of these immune modulators, potentially altering the tumour microenvironment in a favorable way [165].  Thus, Trabectedin appears to affect the biological activity of FUS-CHOP and so far shows promise as a therapeutic in MLPS.5.5. Receptor Tyrosine Kinase Pathway InhibitorsThe high frequency of PIK3CA and PTEN mutations suggests a role for PI3K inhibitors in MLPS. The nonisoform-specific PI3K inhibitors Wortmannin, and LY294002 have been widely used in biological research but are not particularly suited to clinical work due to their lack of specificity, Wortmannin’s instability and LY294002’s low potency (as reviewed [166]). GDC-0941 and PX-866 are promising PI3K inhibitors currently in clinical trials that have low nanomolar potency against class I isoforms of PI3K [167–169]. In lung cancer cell lines and xenograft models, PIK3CA mutants are more sensitive to GDC-0941 [170]. Similarly, PIK3CA mutant and PTEN-null tumours were sensitive to PX-866 in xenograft models, and phase I clinical trials for solid tumours are currently underway [169]. The Rapamycin derivate Everolimus inhibits the mTOR complex-1 (mTORC1), which is a downstream effector of PI3K. Both H1047R and E545K PI3K mutant cells are sensitive to Everolimus [171]. PIK3CA mutated MLPS represents an ideal candidate for PI3K inhibition.As MET is activated in MLPS and there are many MET pathway inhibitors currently in development and in clinical trials (as reviewed in [172], MLPS may be a good candidate for MET inhibition. For example, the novel and promising inhibitor Foretinib (XL880) inhibits multiple kinases including both MET and VEGFR2 and exhibits extensive biological activity and clinical efficacy in an early Phase I clinical trial in metastatic or unresectable solid tumours [173].6. ConclusionMolecular-based therapeutics are not routinely used in liposarcoma, where surgery, radiotherapy, and chemotherapy remain the mainstay of treatment. Translation of targeted molecular therapeutics in sarcoma has been successfully demonstrated with Imatinib mesylate therapy in c-Kit positive gastrointestinal stromal tumour (GIST) [174]. A major challenge with the use of molecularly targeted therapeutics is to translate disease control into disease eradication. One strategy to achieve this goal is to combine two or more independent molecularly targeted agents in a disease where all of the targets are relevant. The dependence of  WD/DDLPS on amplification of both MDM2 and CDK4 means that this disease represents an important candidate for combination therapy. Recent studies point towards RTK involvement in MLPS oncogenesis, particularly signaling through the PI3K/Akt pathway. This provides an important avenue for new research due to the large number of clinical trials currently underway that target this pathway. Although not considered a molecularly targeted therapeutic, treatment of MLPS with Trabectedin is currently in late stage clinical trials with promising results. It is hoped that emerging technologies, such as next-generation sequencing, will be fundamental in revealing new molecular targets in liposarcoma. Similarly, advances in drug development should enable improvement of molecular therapies with greater sensitivity, specificity, potency, and limited toxicity. Combining technologies in both areas will allow for efficient clinical translation.Conflicts of InterestNo potential conflict of interest is disclosed.AcknowledgmentsThe authors acknowledge Dr. Maya Kansara and Dale Garsed for reviewing the manuscript and Dr. Catherine Mitchell for reviewing the histopathology. D. M. Thomas is a recipient of a Victorian Cancer Agency clinician scientist fellowship. S. Young is a recipient of an Australian Postgraduate Award. R. Conyers is a recipient of an NHMRC Postgraduate Scholarship. R. Conyers and S. Young contributed equally to this work.A GOOD READING ABOUT SARCOMA


Molecules and Cancer Cells

Now that we know that the cell does not discriminate about what is the category of this molecule invading me as classified by humans, we are ready to suggest some unconventional combinations of chemotherapy drugs:
Nexavar-Metformin for hepatocarcinoma
anti-MEK- calcium channel blocker for K-ras expressing lung cancer
Antibiotic with impact on splicing molecule with the MTOR
(to be continued)

Monday, March 4, 2013

The AML discussion continues!

AML-1 mostly corresponds to the alpha subunit of the CBF and contains the RUNX-1 which specializes in hematologic differentiation of the cell, the other portion already contains among other things regulatory or catalytic molecules facilitating many processes of the cell combination with EVI-1 will control signal pathways and growth factors.
This type of AML, or AML in general, appears to be a disease mostly driven by dysregulation of promoters and regulator genes with global suppression of the NK-kB a nd the cyclins/TGF. 
?role of ANDROGEN, interferon and growth factor in EVI-1 positive AML can be raised as a question.  On top of standard induction chemotherapy of course!  This is where Cyclosporine and Antithymocyte Globulin could still have a role (in EVI-1 AML)

One may wonder why TGF is suppressed?  Maybe to stop the cancerous process to form a mass and stay "fluid"?   That is, in granulocytic sarcoma, TGF would be less suppressed?

*Screening for Cervical cancer through PAP smear is now for women after the age of 21 years old and should be every 3 years for women between 21 and 30 years of age.

*Screening for Cervical cancer through PAP smear is now for women after the age of 21 years old
and should be every 3 years for women between 21 and 30 years of age.
*Like other screening, the rates have been decreasing.  Also cost and over-treatment had been of concern and led to revision of the guidelines.  No comments were made about HPV immunization.
*FDA warning about new batches of counterfeit Bevacizumab put out under the label ALTUZAM 400mg/16
BATCH #B6022B01 expiring sometime this year!
* Mycobacterium Tuberculosis can hide from host immune defense in the Bone Marrow!
*successful Hearing restoration performed in Mice bearing USH1C gene mutation!
(JAMA)

Sunday, March 3, 2013

Smooth Muscle Myosin & Leukemia

1.SMMHC, a Smooth Muscle Myosin related marker of differentiation, but also implying the use of multiple regulatory genes that ultimately depress P53 as a way to decrease repair of DNA and allow leukemia to proceed with proliferation. this marker is  seen in Inv-16 Leukemia.  Through interaction with SMAD3, it affects TGF driven migration of leukemic cells.  It also interfere with ACTA 2, TGFBR-2 AND FBN-1,
THIS SUGGEST THAT BLOCKING MIGRATION AND IMMUNE BETA-2 RECEPTOR OF TGF COULD ADD TO ACUTE EOSINOPHILIC LEUKEMIA.  BLOCKING NUCLEAR INTERNALIZATION OF THE BETA SUBUNIT OF THE CBF WOULD HELP IN THE CBF DRIVEN LEUKEMIA IN GENERAL.


2.HOXD3
" Mutations in this particular gene cause synpolydactyly and Brachydactyly. The product of the mouse Hoxd13 gene plays a role in axial skeleton development ...
remember the role of Anti-VEGF /MEK in people with gene that impairs morphogenesis.  leukemia with this mutation could a get a trial!

3 MEIS, a cofactor to the "soul of AML" E3
remember we discussed the AML is characterized by suppression of NF-kB
that suppression is achieved by this MEIS over-expression.  it is the over expression to 
HOS protein which is
 'The homologue of Slimb (HOS) F-box protein is a receptor of the Skp1-Cullin1-F-box protein (SCF(HOS)) E3 ubiquitin ligase, which mediates ubiquitination and degradation of beta-catenin and the inhibitor of NFkappaB, IkappaB.''

Stability of homologue of Slimb F-box protein is regulated by availability of its substrate.


 You block this stuff, you release NF-kB, you slow down leukemia or at least decrease co-existing infection rates (role in transplant patients).
INCREASING NEED OF A TISSUE BANK IN EL PASO.

With the increased need for cancer genetic studies, there is an increasing need of a Tissue bank in El Paso.
This town is at the border with Mexico has somewhat of an homogeneous population of a minority population which is most of the time under-represented in most clinical trials.  Here, 80% of the population is Hispanic, therefore increasing the chance to have a nice homogenous cohort for clinical trials in a minority population.  We have completed a study on risk factors for Breast cancer as perceived by the local population.  Full Analysis is awaiting funding.  Data is still sitting in the office while we are busy seeing and helping patients to survive.  A recent Collaboration with the UTEP (University of Texas in El Paso) statistics laboratory will relaunch this and maybe we will then be able to publish the full study.
Risk perception here is very much influenced by the local culture and practices centered on "family" and "religion".
The UTEP Border Biomedical Research Center has a clean cell line maintained, but they want to do more.  But we are here in a low income city and easily ignored.  President Obama stopped by last year and the city voted for him, but nothing else has happened much since.  CPRIT gave 0.2% of its cancer research funding to the 4th largest City in Texas, EL PASO, a city of 800,000 people! That is all we get: a glance from Authorities!  El Paso is largely ignored.  El Paso Women had to organize a sit-out in protest in front of the White House last year.  They were basically ignored and dismissed with political promises that have never materialized.

We need a Tissue bank here in El Paso, everybody knows it, but brushes it off!

The city is home to Fort Bliss, undoubtedly the largest military base receiving most likely the largest portion of returning soldiers.  But the medical facilities here are rather modest, fraught with financial issues and a hesitant medical leadership!  Even the needed new Beaumont Hospital construction is tied up in court with contractors fighting over the opportunities instead of being built!

We need a tissue bank, if you know any one who can help....! call 915-307-3354

Random News: MELATONIN and NIH GrantsInfo

RANDOM NEWS

*Melatonin which helps with sleep at 3mg, was tested at 20 mg to see if it could help to stop or delay cachexia.  It was not different then placebo.  Case closed. IT FAILED!

*IT IS ESTIMATED THAT 12000 DEATHS COULD BE PREVENTED WITH AN INSTITUTION OF AN AGGRESSIVE LUNG CANCER SCREENING IN THOSE WITH HISTORY OF 1 PACK/DAY FOR 30 YEARS OF MORE!

*We have shown that in the united states more than 3000 African women could be saved yearly if the Breast cancer paradox (low incidence, but high mortality) was corrected by the institution of a comprehensive prevention program.  Well, things are getting worse as recent studies continue to show a decrease of screening in world of corruption and politics.  At CPRIT, they want Commercialization.  The NIH is heavily political.  Just look at a recent submission response:

Thank you for your e-mail to GrantsInfo at the National Institutes of Health (NIH).  We provide general information about NIH extramural medical and behavioral research, research training programs, and the grant application process.
I suggest you discuss potential funding of your project with the appropriate Scientific/Research staff linked in the announcement’s Section VII:  http://grants.nih.gov/grants/guide/pa-files/PA-11-260.html#_Section_VII._Agency
Regards from GrantsInfo
Web:   http://www.nih.gov
Linking you to NIH Funding

This is an automatic reply when you submit a project
no referral to some review board
they will keep you going here and there
leading nowhere because they have their political friends to award money to. another example, not here go somewhere else!

Dear Dr. Kankonde,

In the table shown below, you will find all currently active NIH funding opportunities related to support of tissue, specimen, and biospecimen banks.  It does not appear that there is any specific funding opportunity that will work for your purposes.  You can search further in the NIH Guide for Grants and Contracts by going to http://grants1.nih.gov/grants/guide/search_guide.htm.  You might also try to connect and collaborate with scientists and pathologists who have mutual interests to find out if your proposed tissue bank might be supported through affiliation with a large research program (e.g., medical center, cancer center, program project, clinical trials network/consortium, special program of research excellence, epidemiology/population cohort, etc.)

Good luck with your efforts.

Comment: and it goes on and on, you get nowhere and after many attempts, you just give up (but at the CRBCM, we do never give up!)

It is a rough world, and they claim to help, but indeed they try to discourage any pursuit of new research and prevention with good old political tactics!

Saturday, March 2, 2013

HYPOTHESIS FOR TARGET THERAPY IN AML

Leukemic cells do not like Mesenchymal transformation, therefore MEK amplification is an absolute must in treatment.
2.  The primary dysfunction in AML is Histone Deacetylation
but Histone deacetyl transferase inhibitors will work in some cases because of the variety of deacetylation or heterogeneity of molecules involved
3.  Amplification of the NF-kB signal transduction may help some patient
4. Growth factors and Androgen  may help some patients
5. Insulin may help some subset of patients
6. we predict that MTOR inhibition will have a very limited effect (except in those with specific gene discussed yesterday)
7. Velcade will not work in patients with NACA-1

so combination of the future
chemoth and MEK amplification agents
Histone Deacetylase inhibitors and MEK amplifiers
Histone Deacetylator Androgen and MEK amplifiers
MTOR-ANDROGEN-MEK amplifier
androgen will be added to pt with TIF2
IN AML FLT3 IS NOT PREDICTIVE OF RESPONSE TO THERAPY, IT HAS ONLY PROGNOSIS VALUE.

Remember we explained that in good prognosis AML, those that involve Core Binding Factor (CBF), the disruption in this molecule appears to involve disruption of histone deacethylation primarily which an activity of the Beta subunit of the CBF and we showed that the NF-kB was suppressed.  There is an associated  significant depression of cyclins and TNF, TGF which are important for CD135 (FLT 3) receptor entrance into the cell for signal transduction flow to occur. when the FLOW of this pathway is going fine, the receptor decrease at the membrane.  But under lack of TGF, the membrane FLT3 receptor transfer into the cell or its use is decreased causing Mis-location (a well known cellular dysfunction phenomena). Too many FLT-3 lead of course to dimerization and polymerization forming "Tandem" or repeats of FLT-3.
Now FLT3 suppression by growth factor is part of the normal differentiation of cell, or aging of Hematopoietic cells.   Overexpression of the FLT3 as a result confers to the cell dediffentiation and stem cell like characteristics.
The answer of course is now to increase TGF which may not work because Nuclear substrates are already compromised in most cases.  Increase Mesenchymal transformation through MEK is the only option which secondarily will increase VEGF.  Regorafenib provide here the best alternative even though the Sutent have shown activity.  Anti-MEK is the only significant intervention in AML.
Now don't get yourself cocky.  Remember the primary event is Nuclear, so anti-MEK alone cannot solve the issue and could only be for elderly. Give Anti-MEK with the standard Induction regimen and barace yourself for a new panel of side effects.

Another option histone Deacetyl transferase inhibitor and anti-MEK, this may be the chemoth free option!
anti-MEK and Arsenic Trioxide in APL!
Target therapy is here, let's use it!
TIF 2: TRANSCRIPTION INTERMEDIATE FACTOR 2
-------------------------------------------------------------A nuclear protein complex containing a Histone deacethyl transferase and a receptor to steroids which attachs Glucocorticoid, Estrogen (BRCA-1)and facilitate DNA transcription.
Also known as Nuclear Receptor Coactivator2 NCOA-2
Overexpression of TIF or mutaion here may have an impact on one of the subunit of Cytochrome-C,
It is a ligand (other ligand SRC-1, AIB-1).  It may help in tolerance of certain stressful milieu!
Is TIF 2 a biomarker for steroid response?
does TIF2 Mutation compromise effectiveness of ATRA in APL?
what the role or interaction with NACA-1?
how does PIF2 intervene in aging of cells?
what is the role of Androgen in AML expressing TIF-2?

ACTIVITY AT CRBCM.

1. WE WON ANOTHER SMALL CONTRACT, WILL WORK FROM INDIANAPOLIS, IN FOR 4 WEEKS. SHIFTING BACK AND FORTH AS NEEDED. GOOD THING EL PASO AIRPORT HAS BEEN EXTENDED TO GO INTO FORT BLISS. PLENTY OF ROOM TO LAND!  CRBCM STAYS ALIVE!

2.STILL FIGHTING FOR FUNDING, SUBMITTED 2 NEW GRANT APPLICATIONS BEFORE THE NIH DESPITE OUR PREVIOUS NEGATIVE EXPERIENCE.

3. FINALLY AFTER 10 MONTHS THE IRS CAME ALIVE TO ADVANCE TAX EXEMPTION FILE FOR THE CRBCM.  THEY NEED MORE INFORMATION.  THIS ORGANIZATION HAS COST US 2 SUPERB OPPORTUNITIES TO WIN A GRANT.  NOW I BELIEVE THE REPUBLICANS WHEN THEY SAY THAT A BIG GOVERNMENT IS BAD FOR YOU!  AND THEY STILL HAVE NOT PROCESSED OUR TAX RETURN SUBMITTED 4 WEEKS AGO.

4.MEDICARE AND MEDICAID REIMBURSEMENT STINKS.  PEOPLE TALKING ABOUT SEQUESTER AND PAY CUTS TO DOCTORS ARE MISGUIDED. THE GOVERNMENT HAS STOPPED PAYMENT ALL TOGETHER. DOCTORS ARE NO LONGER TAKING MEDICAID PATIENTS BECAUSE THE GOVERNMENT HAS STOPPED PAYMENTS OR SITS ON PAYMENTS FOR 180 DAYS, THAT IS 6 MONTHS! WE ARE NOW ASKING PATIENTS TO PAY UPFRONT OR ELSE!

5. GOOD TO KNOW THAT THERE ARE PRIVATE PLANES TO FLY CANCER PATIENTS FOR FREE TO MD ANDERSON FROM EL PASO.  THERE ARE STILL GOOD SOULS TO THE EL PASO PATIENTS SINCE THE GOVERNMENT HAS FAILED ONCE AGAIN!  WE WORK HARD HELPING PATIENTS GET THERE!  IT  IS PATIENT NAVIGATION AT WORK!

6. THE VETERANS ADMINISTRATION AND THE MILITARY IN EL PASO HAVE NO INCLINE FOR RESEARCH, NO LAB FACILITY AND THEIR LEADERS HAVE NO IDEA OR CONSIDERATION FOR SUCH "RISKY "ADVENTURE.  WASTING LARGE OPPORTUNITIES OF FUNDING FROM THE DEPARTMENT OF DEFENSE.  THIS COMMUNITY WHICH HAS ONE OF THE LARGEST AND EXPANDING GROUPS OF RETURNING SOLDIERS COULD HAVE ATTRACTED FUNDING FROM DOD.  BUT THEIR LEADERS ARE JUST ANOTHER BUNCH OF PAPER PUSHERS WAITING FOR SAFE RETIREMENT.  FOR PEOPLE WHO ARE FINANCIALLY STRAPPED, YES MEDICAL FACILITY OPERATIONS AT FORT BLISS ARE "IN THE RED", THEY ARE NOT CREATIVE!  THEY TOTALLY IGNORE DOD FUNDING RESOURCES!  CONGRATULATION FOR A SOLID MILITARY PERFORMANCE! WE JUST NEED NEW LEADERS AT FOR BLISS AND VETERANS AFFAIRS IN EL PASO!

Friday, March 1, 2013

TB Advisory: Nationwide Shortage of Tuberculin

The CDC received the following statement from Sanofi-Pasteur yesterday regarding a supply interruption for Tubersol®.  Current demand on the 10 test presentation size has placed the company in a position where it perceives that it will temporarily need to allocate the test. 
TUBERSOL is manufactured in 2 presentation sizes: 10 tests per presentation and 50 tests per presentation. TUBERSOL Diagnostic Antigen in the 50-test presentation is experiencing a supply interruption until late-spring 2013 due to a temporary delay in production. As a result, increased demand has limited the availability of TUBERSOL Diagnostic Antigen in the 10-test presentation.  To allow us to continue to supply the market with product, we are placing allocations on customer orders which will go into effect immediately.  Allocations will be based on a percentage of historical usage for customers who order an average of 10 doses or more per month, and these allocations are expected to be in effect until late-March. A follow-up communication will be sent in late-March when we expect to lift these allocations. –Sanofi-Pasteur
Indiana State Department of Health’s TB Program recommends the following measures until the shortage is resolved.
 
Recommendations for Responding to Tuberculin Shortages
 
·         If available and appropriate, screen for LTBI with an Interferon Gamma Release Assay (IGRA) (T-SPOT®.TB and QuantiFERON® Gold in-tube) instead of a tuberculin skin test (TST).
·         Prioritize TSTs if necessary.  High priority groups include:
o   Contacts to a person with pulmonary or laryngeal TB
o   Persons who are immuno-compromised
o   Evaluation of persons with symptoms suggestive of TB disease
·         If necessary, defer annual screening of employees, residents, and/or inmates as part of an infection control plan until sufficient tuberculin becomes available.
 
 
Aplisol®, an alternative formulation of tuberculin, is an acceptable alternative to Tubersol® for tuberculin skin testing. It is recommended that programs that do serial testing use the same preparation of tuberculin year to year to avoid reported issues with false-positive tests associated with switching preparations. Aplisol® is a good choice for persons requiring one-time screening or for testing performed as part of a contact investigation (i.e. initial and 8-week testing) as long as Aplisol® is used for both tests. Be advised that due to anticipated increased demand, it is not clear that an adequate supply of Aplisol® will be available to meet all future need.[1]
Questions and concerns about this advisory may be addressed to Sarah Burkholder, RN, MPH, Director of the TB/Refugee Health Division at 317-233-7545 or sburkholder@isdh.in.gov.  
____________________________________________
Physicians mailing list
Physicians@lists.in.gov
http://lists.in.gov/mailman/listinfo/physicians
PART III AML
=================
WE WERE SAYING:

Should CBF activity lead to MYCN amplification directly or through overexpression of miR17-92 (with ribosomal location), at least 8 consequences results:

1.Overexpression of CDK4  (Cyclin dependent kinase 4) which phosphorylate RB-1 and stops the cell cycle in G-1.  Remember p16ink 4a is its inhibitor (Melanoma).   Therefore MYCN stops proliferation!  That's why its a good prognostic factor!

2.  Overexpression of RSG2 ( G protein regulator) mostly with inhibitory membrane and submembrane activity effects affecting cell division  as well as angiogenesis, again good for you!

3.Overexpression of MDM2, a ubiquitination ligase of P53.  In Leukemia, proliferation will be exacerbated by this action (not so good for us! but inhibitors could prove good target therapy) 
   
4. Overexpression of TERT, This is where the AML cells insure longevity by exacerbating  or mutating the the Telomerase Reverse Transcriptase, insuring the stability of DNA repeats or prolonging the tail that protect chromosome-This is not food for us.  Association with Hypoxia induced factor point more likely to preparation to handle stress and possibly tissue invasion that occur sometimes with leukemia.

5.Overexpression/Mutation of RNF11 This gene seems to code for a Ligase that allows protein-protein interaction.  Ubiquitination and attachment to membrane protein may be compromised limiting full deployment of NF-kB pathways.  it may account for  susceptibility to infection as cellular immunity would be severely compromised.  Sometime frame shift Mutation is all it takes when miRNA are targeted.
(we will look into the role of DUMPS and CD-38 as they pertain to RNF11)
-----------------------------------------------------------------------
6.  Interaction with - E2F1, a transcripton factor,  through inhibtion of Rb-1, controls proliferation, 

                             - ODC-1, RATE limiting enzyme at the base of the amplification system and therefore at the center on Oncogenesis, you knock down the OC-1, good bye amplification since mutation here removes the rate limiting effects  (sensitive target!)  (Ornithin Carboxylase 1)
                        
                              -PPM1G
Roni Koren at al wrote:
"PP2A1 is one of the major protein serine/threonine phosphatases in the cell, which plays a role in several cellular processes, including metabolism, transcription, RNA splicing, translation, cell cycle progression, morphogenesis, signal transduction, development, and transformation (1, 2)."

PPM1 mutation is the soul of Dedifferentiation of Myeloid cells, and possible increase of migration.
"Mutations in PPM1, which methylates protein phosphatase 2A, and target of rapamycin (TOR1) were characterized further. Deletion of PPM1 almost completely suppressed the rapid lethality and substantially suppressed glucose wasting during starvation for leucine or uracil. Suppression by a deletion of TOR1 was less complete."
Viktor  Boer at al reported. This hinted that PPM1 Mutation presence may mitigate MTOR inhibition effect.

These 3 alterations involve Chromosome 2
------------------------------------------------------------------------------------
7.   Over-expression of INHBE, NACA, SHMT2

-INHBE Better described here
 

UniProtKB/Swiss-Prot: INHBE_HUMAN, P58166

Function: Inhibins and activins inhibit and activate, respectively, the secretion of follitropin by the pituitary
gland. Inhibins/activins are involved in regulating a number of diverse functions such as hypothalamic and pituitary
hormone secretion, gonadal hormone secretion, germ cell development and maturation, erythroid differentiation, insulin
secretion, nerve cell survival, embryonic axial development or bone growth, depending on their subunit composition.
Inhibins appear to oppose the functions of activins


-NACA
 " NAC binds to nascent proteins as they emerge from the ribosome, blocking interaction with the signal recognition particle (SRP) and preventing mistranslocation to the endoplasmic reticulum. However, nascent proteins with an exposed signal peptide will not be bound by the encoded protein, enabling them to bind the SRP and enter the secretory pathway. This protein has been determined to be an IgE autoantigen in atopic dermatitis patients. Several transcript variants encoding two different isoforms have been found for this gene. [provided by RefSeq, Oct 2011]"

-SHMT2
' Serine hydroxymethyltransferase (SHMT) is an enzyme (EC 2.1.2.1) which plays an important role in cellular one-carbon pathways by catalyzing the reversible, simultaneous conversions of L-serine to glycine (retro-aldol cleavage) and tetrahydrofolate to 5,10-methylenetetrahydrofolate (hydrolysis).[1] This reaction provides the largest part of the one-carbon units available to the cell.[2]' wikipedia



 Globally, leukemic cells seems to stay dedifferentiated  by shielding themselves away from mesenchymal transformation and away from the effect of growth factors.  the SHMT2 challenge us to redefine the role or not of anti-folate.  Here we know where one carbon-Unit are coming from.  We also have here futher evidence that the NF-kB is completely dysrgulated or suppressed in Leukemia.  DKK3 SUPPRESSION PUT SUPPRESSION OF THE WNT SIGNAL TRANDUCTION CENTER TO THE LEUKEMIC PROCESS.  DEREPRESSION HERE IS NEEDED.




8.  Suppression of DKK3, ESR1, TGF beta 

DKK3 
"is involved in embryonic development through its interactions with the Wnt signaling pathway. The expression of this gene is decreased in a variety of cancer cell lines and it may function as a tumor suppressor gene. Alternative splicing results in multiple transcript variants encoding the same protein.[2]"wikipedia 

DKK3 SUPPRESSION PUT SUPPRESSION OF THE WNT SIGNAL TRANDUCTION CENTER TO THE LEUKEMIC PROCESS.  DEREPRESSION HERE IS NEEDED.





ESR 1

DECREASE OF ESTROGEN 
ALPHA RECEPTOR IS USED HERE TO RESTRAIN DIFFERENTIATION AND ALLOW GROWTH UNDER STRESSFUL CONDITION, AND CONTROL AGING.

TGF1 IS ALSO SUPPRESSED TO AVOID GROWTH AND DIFFERENTIATION


GLOBALLY IT APPEARS THAT DIFFERENTIATION THROUGH MEK, PITUITARY HORMONES, GROWTH FACTOR (TGF beta), INSULIN, COULD HAVE A SHOT AT MINIMIZING ACUTE LEUKEMIA BASED ON INTUITIVE DATA PRESENTED ABOVE!  IS LEUKEMIA RATES LOW IN DIABETICS WHO ARE TAKING INSULIN?
PROF OF CONCEPT NEEDED AT CRBCM.  INCREASING OR PROTECTING WNT SIGNALING MAY BE ANOTHER FIGHTING APPROACH. 
MEDICATION BUST, THE CASE OF CILENGITIDE IN TREATMENT OF GLIOBLASTOMA.

Once again we have seen a failure of a medication that was ill used.  It was used on the simple premise that in Gliomas, the cell over expressed certain integrins.  Therefore an inhibitor of "integrins" would work in this challenging disease.  The Integrins discussed were just as fuzzy.  They seemed to involve cell migration more than angiogenesis, in a disease which does not metastase.  Suggestions of anti-FAK, anti-Src, anti-AKT activities have been made peripherally but the emphasis is that this drug would meet the challenge of some integrins which could be more prognostic than predictive.  These integrins appear to have more diagnostic than predictive.  You need to go after a driver mutation or a significant secondarily amplified mechanism.
In Glioblastoma, the role of NF-1, VGEF, MEK, Interferon Receptor seems to be more of importance.  Integrins involved in metastasis in  Glioma? Really?
When you go after SRC, pick carefully which SRC!
It's like when you go after RAS, pick the right RAS!
When you go after MYC, pick the right MYC !
Globally when you go after differentiation, be selective or go after MEK, the door to differentiation in adult. It is not by mistake that people who where going after K-RAS as a marker went in with an anti-MEK in lung cancer!

If I had to pick a role for Cilengitide, I would use it with Avastin or an anti-MEK
and I would use it in Maintenance setting at least from info I had gathered on it!  And in Glioma, in 2nd line where Avastin is king!

I will here finish by just saying if someone says "anti-Integrin", he has given you no information whatsoever, how many integrins are in a cell.  If you find someone who gives a straight answer, call me please!