Rev 1500 Schedule B: Fill & Download for Free

GET FORM

Download the form

How to Edit Your Rev 1500 Schedule B Online On the Fly

Follow the step-by-step guide to get your Rev 1500 Schedule B edited with efficiency and effectiveness:

  • Hit the Get Form button on this page.
  • You will go to our PDF editor.
  • Make some changes to your document, like highlighting, blackout, and other tools in the top toolbar.
  • Hit the Download button and download your all-set document into you local computer.
Get Form

Download the form

We Are Proud of Letting You Edit Rev 1500 Schedule B Like Using Magics

Get Our Best PDF Editor for Rev 1500 Schedule B

Get Form

Download the form

How to Edit Your Rev 1500 Schedule B Online

If you need to sign a document, you may need to add text, complete the date, and do other editing. CocoDoc makes it very easy to edit your form with the handy design. Let's see the easy steps.

  • Hit the Get Form button on this page.
  • You will go to our PDF text editor.
  • When the editor appears, click the tool icon in the top toolbar to edit your form, like signing and erasing.
  • To add date, click the Date icon, hold and drag the generated date to the target place.
  • Change the default date by changing the default to another date in the box.
  • Click OK to save your edits and click the Download button once the form is ready.

How to Edit Text for Your Rev 1500 Schedule B with Adobe DC on Windows

Adobe DC on Windows is a useful tool to edit your file on a PC. This is especially useful when you finish the job about file edit without using a browser. So, let'get started.

  • Click the Adobe DC app on Windows.
  • Find and click the Edit PDF tool.
  • Click the Select a File button and select a file from you computer.
  • Click a text box to modify the text font, size, and other formats.
  • Select File > Save or File > Save As to confirm the edit to your Rev 1500 Schedule B.

How to Edit Your Rev 1500 Schedule B With Adobe Dc on Mac

  • Select a file on you computer and Open it with the Adobe DC for Mac.
  • Navigate to and click Edit PDF from the right position.
  • Edit your form as needed by selecting the tool from the top toolbar.
  • Click the Fill & Sign tool and select the Sign icon in the top toolbar to customize your signature in different ways.
  • Select File > Save to save the changed file.

How to Edit your Rev 1500 Schedule B from G Suite with CocoDoc

Like using G Suite for your work to complete a form? You can make changes to you form in Google Drive with CocoDoc, so you can fill out your PDF without worrying about the increased workload.

  • Go to Google Workspace Marketplace, search and install CocoDoc for Google Drive add-on.
  • Go to the Drive, find and right click the form and select Open With.
  • Select the CocoDoc PDF option, and allow your Google account to integrate into CocoDoc in the popup windows.
  • Choose the PDF Editor option to open the CocoDoc PDF editor.
  • Click the tool in the top toolbar to edit your Rev 1500 Schedule B on the needed position, like signing and adding text.
  • Click the Download button to save your form.

PDF Editor FAQ

Which is older language, Hindi or Marathi?

To get to know which one is older we need to get to the origin of each language. Marathi and Hindi both are Sanskritized versions of Prakrit languages and are written in the same script in recent period (1850 onwards).Origin of HindiAll of the languages of India can be grouped into two major group that are 1) Indo-European languages that include Hindi, Punjabi, English, German, Spanish, Bengali etc. 2) Dravidian Languages that includes Tamil, Telugu, Malayalam and KannadaIndo-Aryan LanguagesThe Indo-Aryan languages is a branch of the Indo-Iranian languages which is in itself a branch of the Indo-European languages group. Indo-Aryan languages has a base of about speakers form about 1.5 billion of whole 3 billion Indo-European speakers. This sub-group also account for more than half of all recognized Indo-European languages. The largest native speakers in this group belongs to Hindustani (Standard Hindi and Urdu), Bengali, Punjabi, Marathi , Gujarati, Oriya, Sindhi, Nepali, Sinhala etc.In Indo-Aryan language group, the oldest language is Vedic Sanskrit (1500 BC – 800 BC). Vedic Sanskrit is an archaic form of Sanskrit and it is the oldest attested language of the Indo-Iranian branch of the Indo-European family. This form of Sanskrit is the language of ancient Indian scriptures Vedas. Vedic Sanskrit was the origin of languages which later gave birth to Hindi and other languages in this group.After Vedic Sanskrit, another form of Sanskrit came into growth path. It wasClassical Sanskrit (800 BC – 500 BC), which was the language of nobles and influential upper class. Classical Sanskrit is still one of the 22 scheduled languages of India and is an official language of the state of Uttarakhand. In present time, the speakers of this language are almost negligible, it is still taught in Indian school in effort to preserve it like Latin in western societies.Classical Sanskrit was language of upper class however there was another language, Prakrit (500 BC – 500 AD) which had directly developed from Vedic Sanskrit in the same era. It was vernacular of common mass and it was developed in natural way in contrast to the literary and religious language, Classical Sanskrit. During the era, another form of Prakrit, Pali (500 BC – 500 AD) was in use. It was a literary language of the Prakrit language family. It is mostly seen in Buddhist scriptures. Pali was also used extensively by a legendary emperor of India, Asoka the Great (304 BC – 232 BC).Both Prakrit & Pali, continue to grow in vernacular and gave birth to another language, Apbrunsh ( 500 AD – 1000 AD). Apbrunsh later gave way to Khari Boli (900 AD – 1200 AD). It was developed in the north Indian part.From Khari Boli, a more refined language Hindustani which was a mix of present day Hindi and Urdu came into use. In 19th century, both Urdu and Hindi has separate and refined in their own way. Hindi and Urdu share much of the grammar though both used different script for writing and Hindi uses more of its word which are derived word from Sanskrit. Urdu derives more of its derived word from Persian, Arabic and Turkish.Origin of MarathiMarathi is the language of more than fifty million people mostly residing in Maharashtra, the region in western India with Mumbai as its capital. However, the name Maharashtra does not occur in the Ramayana, nor in the Mahabharata. The Chinese traveler Yuan-Chwang referred to this area, in the seventh century as Mo-ha-la-cho. In tenth century Al Beruni mentions the Marhatta region with Thane as its capital. Till then Konkana was not included in this area; Soparak was its other name (modern Sopara, the harbour).There is no unanimity amongst scholars about the origin and antiquity of this language. The first written form is in Vijayaditya's Copper-plate, dated 739 A.D., found in Satara. In 983 A.D., the stone inscription at the feet of Shravanabelgola Gomateshwar- Chavundarajen Karaviyalen (Built by Chavandaraja, the king), is considered to be the oldest. An interesting couplet in the Jain monk Udyotan Suri's Kuvalayamala in the eighth century, refers to a bazar where different people speak differently, selling their goods: the Marhattes speak Dinnale, Gahille (given, taken).About the genealogy of the language scholars have different views: C.V. Vaidya maintained that it developed from Sanskrit, Sten Knonow maintained that it developed from Maharashtri Apabhramsa, others regard it as one of the Pancha Dravida (five Dravidian) groups. Khaire has recently found several loanwords in early and spoken Marathi from Tamil (adgule-madzule). There were many borrowings from Telugu (tup, tale) and Kannada words are the highest in Marathi spoken under Yadavas (1180-1320). In 1290A.D. the Hoyasala minister Perusmala at Mailangi made ``provision for masters to teach Nagar, Kannada, Telugu and Marathi'' (B. Lewis Rice in Mysore and Coorg from inscriptions) Later Marathi, in Shivaji's times imbibed many words from Persian, Arabic, Portugese and English Maharashtra Shabdakosh (in eight volumes edited by Y.G. Date and C.G. Karve) has 1,12,189 words, out of which the words from Persion-Arabic stock are 2,900 and from European stock 1,500. The script used for Marathi writing is the same as Devanagari, with an additional ``L'', old Marathi historical documents are found in Modi script. In 1622, Father Stephans wrote Khrista-Purana in chaste Marathi. The language was enriched by several writers who were Muslims (like saint Sheikh Muhammad, the Sufi) or Rev. N.V. Tilak (1865-1919).So We can say that the Marathi is older language than Hindi according to available history of the languages and their origin.Sources :Central Hindi DirectorateControversies in HistoryHindi Language BlogThanks for A2A.

What cancers are associated with infectious diseases?

About 15% of human cancer are infection related.See...The Yale Journal of Biology and MedicineYale Journal of Biology and MedicineViruses and Human CancerJohn B LiaoAdditional article informationAn estimated 15 percent of all human cancers worldwide may be attributed to viruses [1], representing a significant portion of the global cancer burden. Both DNA and RNA viruses have been shown to be capable of causing cancer in humans. Epstein-Barr virus, human papilloma virus, hepatitis B virus, and human herpes virus-8 are the four DNA viruses that are capable of causing the development of human cancers. Human T lymphotrophic virus type 1 and hepatitis C viruses are the two RNA viruses that contribute to human cancers.Close study of viruses and human cancer has led to optimism for the development of new strategies for the prevention of the preceding infection that can lead to carcinogenesis. The presence of viral gene products in tumor cells that require them to maintain their unchecked proliferation also can provide important targets for directed therapies that specifically can distinguish tumor cells from normal cells. The inability of traditional cancer therapy, such as chemotherapy and radiation, to distinguish cancer cells from normal cells is a significant drawback and leads to toxicities for patients undergoing treatment. Targeted therapies directed against viral proteins or generate immune responses in order to either prevent infection or kill infected cells or cancer cells hold much promise for more effective and tolerable strategies.Human tumor virusesAlthough it is convenient to consider human tumor viruses as a discrete group of viruses, these six viruses, in fact, have very different genomes, life cycles, and represent a number of virus families. The path from viral infection to tumorgenesis is slow and inefficient; only a minority of infected individuals progress to cancer, usually years or even decades after primary infection. Virus infection also is generally not sufficient for cancer, and additional events and host factors, such as immunosuppression, somatic mutations, genetic predisposition, and exposure to carcinogens must also play a role.Hepatitis B and C virusesHepatitis C virus is an enveloped RNA virus of the flavivirus family. It is capable of causing both acute and chronic hepatitis in humans by infecting liver cells. It is estimated that approximately 3 percent of the world’s population are hepatitis C carriers [2]. Chronic infection with hepatitis C virus results in cirrhosis, which in turn can lead to primary hepatocellular carcinoma. Between 1 and 2 percent of infected patients with subsequent compensated cirrhosis will develop primary hepatocellular carcinoma per year [3]. Transmission of the virus occurs through the blood, with shared needles in intravenous drug abuse, sexual activity, and parturition being the primary routes.The hepatitis B virus of the family hepadnaviridae is, by contrast, a DNA virus, but the features of its resulting disease share many similarities with hepatitis C virus. Hepatitis B virus also is a blood-borne pathogen that can result in acute and chronic hepatitis. Chronic hepatitis, that is, infections lasting more than three months, can lead to cirrhosis and liver failure. Chronic infection also can lead to the development of hepatocellular carcinoma [4]. Hepatitis B infections is a significant global health problem with an estimated 2 billion people infected and 1.2 million deaths per year attributed to subsequent hepatitis, cirrhosis and hepatocellular carcinoma [5].Hepatocellular carcinoma is an aggressive tumor that can occur in the setting of liver disease resulting from infections with hepatitis B and/or hepatitis C virus, although the exact mechanism of oncogenesis by these viruses is unclear. Diagnosis is usually made late in the course of liver disease and median survival ranges from six to 20 months after that time [6]. The traditional foundation of treatment is surgical, whether tumor resection or transplantation. However, nonsurgical options such as percutaneous ethanol injection, transarterial embolization, radiofrequency ablation, chemotherapy, and radiotherapy are also utilized. The choice of therapies frequently depends on the extent of disease and the amount of liver function the patient has in reserve [7,8].Research into novel therapies have focused on the use of virally targeted and immunological strategies with an eye on preventing infection. Unfortunately, hepatitis C virus has proved to be poorly suited to vaccines because its genome possesses a very high mutation rate, especially in the hypervariable region of the genome coding for the envelope proteins allowing it to escape immune recognition and elimination by the host. There are 11 distinct genotypes and several subtypes identified.The introduction of vaccines against hepatitis B virus in the early 1980s marked a major milestone with what might be considered the first cancer prevention vaccine, although the primary goal of this vaccine was to prevent hepatitis. Since that time, more than 110 countries have adopted a universal policy of immunizing all newborns, according to the World Health Organzation. Additionally, countries that have successfully implemented this program significantly have decreased the carrier rate and infection in their populations [9]. However, vaccine coverage is often low in many developing countries due to the cost, lack of heath care infrastructure for delivery of the vaccine, and the need for three needle injections over six months. Even in some developed nations, universal vaccination has not been implemented because of the belief that it is a limited public health problem and the expense is not justified [10,11].New challenges for combating hepatitis B infection center around efforts to address the limitations of the current vaccine: the need for multiple injections, the presence of up to 10 percent nonresponders to the vaccine, the discovery of hepatitis B virus S gene escape mutants in infants that were infected despite an adequate response to the vaccine, and the cost for developing nations. The current multiple dosing schedule is being addressed with attempts to combine it with other required vaccines or decrease the number of doses. Oral vaccination also is being investigated as a way to obviate the need for trained personnel to administer injections. The World Health Organization estimates that from $8 to $12 billion will be needed to immunize children from the poorest countries from 2005-2010, which has prompted efforts from public and private organizations to advocate for funding to fill the need.Medical therapy for patients infected with hepatitis B has focused on the use of interferon to reduce viral replication, which decreases the incidence of life-threatening liver complications in patients who respond to the treatment [12]. Interferon alpha treatment is effective in 20 to 30 percent of cases in inducing loss of the hepatitis B e antigen. However, the impact of interferon therapy on subsequent hepatocellular carcinoma rates is less clear [13,14]. Interferon therapy is also limited by cost and side effects.The limitations of interferon therapy have been partly circumvented with the use of targeted antiviral agents. Lamivudine has been shown in a large multicenter randomized placebo-controlled trial to be effective in reducing both the incidence of hepatic decompensation and the risk of hepatocellular carcinoma [15]. Other antiviral agents continue to join the armamentarium; lamivudine, adefovir, entecavir, and telbivudine have been shown to be effective in hepatitis B disease. These agents are nucleotide analogues that exploit the need for the hepatitis B virus to use reverse transcriptase to replicate viral DNA. Since these agents specifically target the viral replication machinery and are given orally, they are better tolerated. However, it has been observed that long-term therapy with lamivudine can lead to the emergence of genotypic resistance mutations [16], but this does not negate the benefits of lamivudine therapy in reducing the rates of hepatocellular carcinoma [15]. The success of these therapies has reached the point where patients with advanced cirrhosis secondary to hepatitis B can be treated and transplanted without the development of hepatitis B in the transplanted liver.Medical treatment of infection with hepatitis C has not progressed at the same speed. Pegylated interferon with ribavirin, an antiviral agent that may act as a nucleoside analogue and inhibitor of RNA dependent RNA polymerase, has been shown to be successful in eradicating infection in half of patients [17]. However, therapy is expensive and side effects are significant. Phase II trials of oral antivirals such as protease inhibitors and polymerase inhibitors are currently under way [18]. Unlike hepatitis B, treatment of hepatocellular carcinoma due to hepatitis C infection with transplantation almost always results in recurrent infection of the transplanted liver [19].The search for targeted therapies that can block hepatitis C viral replication by selectively inhibiting viral replication has for many years been hampered by the lack of experimental infection systems, in either cell culture or animal models to test candidate therapies. The recent development of viral replicons, subgenomic RNAs that are expressed and autonomously replicate within cells, has led to the use of hepatitis C viral replicons that can replicate in human hepatoma cells lines [20] and the development of mouse models of the disease [21,22]. These advances may herald more rapid progress in the development of virally targeted therapies such as hepatitis C virus specific protease and polymerase inhibitors.Epstein-Barr virus (EBV) and human herpesvirus 8 (HHV-8)EBV and HHV-8 (also known as Kaposi sarcoma herpesvirus) are both herpesviruses that possess large double-stranded DNA genomes. As with all herpesviruses, they encode enzymes involved in DNA replication and repair and nucleotide biosynthesis. They also both possess the ability to establish latency in B lymphocytes and reactivate into the lytic cycle. Both also are associated with naturally occurring tumors in humans.EBV is a ubiquitous virus that is most commonly known for being the primary agent for infectious mononucleosis. Up to 95 percent of all adults are estimated to be seropositive, and most EBV infections are subclinical. EBV also is associated with a number of malignancies: B and T cell lymphomas, Hodgkin’s disease, post-transplant lymphoproliferative disease, leiomyosarcomas, and nasopharyngeal carcinomas. Of these cancers, Burkitt’s lymphoma, post-transplant lymphoproliferative disease, and leiomyosarcomas show an increased frequency in patients with immunodeficiency, suggesting a role for immunosurveillance in the suppression of malignant transformation.The primary site of infection is the oropharyngeal cavity, and EBV is capable of infecting both B cells and epithelial cells and switching between the two [23]. The major surface glycoprotein, gp350/220, binds to the cd21 receptor on B cells. Transformation of B cells is a highly efficient process requiring a large portion of the EBV genome, which becomes circular for replication and latency. Virus will directly enter the latent gene expression state with suppression of the lytic cycle. Production of a number of latent gene products are required for immortaliztion.Immune therapy of EBV-associated tumors has been target of research since standard therapy generally has entailed the use of multi-agent chemotherapy, radiation therapy, and surgery. This work has centered around adoptive transfer of EBV-specific cytotoxic T-cells [24,25] and shown success but must overcome obstacles such as potential graft vs. host disease and resistance due to mutation of selected EBV epitopes [26]. Vaccines capable of preventing primary EBV infection or boosting immune responses against EBV-associated tumors are under investigation. Much of the development thus far has focused on gp350/220 subunit vaccines [27], since it is one of the most abundant proteins on the virus coat and also the protein against which the human EBV neutralizing antibody response is directed [28]. Another strategy involves the use of a recombinant vaccinia viral vector to express an EBV membrane antigen [29]. A successful vaccine would have the greatest impact in regions of the world that have an especially high incidence of specific malignancies. Burkitt’s lymphoma is the most common childhood malignancy in the central part of Africa where EBV and malaria are considered cofactors in its carcinogenesis and 95 percent of children are infected by age 3, compared to the United States, where infection is usually delayed until adolescence [30]. Nasopharyngeal carcinoma is relatively rare but has an exceptionally high incidence in southern China, approaching more than 20 times greater than that of most populations [31].In 1994, HHV-8 DNA was identified in biopsies from tumors of a patient with Kaposi sarcoma [32], a relatively rare malignancy prior to the AIDS epidemic. In addition to it likely being an essential cofactor for the development of Kaposi sarcoma, HHV-8 also is believed to have a role in Castleman’s disease and primary effusion lymphoma [33]. The viral genome is expressed in these tumors and encodes transforming proteins and anti-apoptotic factors. The virus is also able to enhance the proliferation of microvascular endothelial cells [34]. As with EBV, the predominant infected cell is the B lymphocyte, although here the lytic cycle is embraced rather than repressed. This may play a crucial role in the pathogenesis of Kaposis sarcoma by elaboration of viral and host cytokines promoting cell proliferation, angiogenesis, and enhancement of viral spread.Targeted antiviral agents such as ganciclovir directed against viral DNA replication have had a dramatic affect on decreasing the incidence of Kaposi sarcoma in AIDS patients through both therapy and prophylaxis [35]. Ganciclovir is phosphorylated into a GTP analog, which acts as a competive inhibitor of viral DNA polymerase resulting in termination of viral DNA elongation. Furthermore, a G protein coupled receptor (vGPCR) has been identified as a viral oncogene in HHV-8 infected cells that can exploit cell signaling pathways to induce transformation and angiogenesis [36]. vGPCR also has been proposed as a target for novel molecular therapies because of its key role in disease progression [37]. But the therapy regimen most responsible for the decreasing incidence of Kaposi sarcoma may well be the success of highly active antiretroviral therapy (HAART) regimens targeting HIV [38], since it was the emergence of HIV that led to the increasing incidence of Kaposi sarcoma.Human Papillomavirus (HPV)HPV are small non-enveloped DNA tumor viruses that commonly cause benign papillomas or warts in humans. Persistent infection with high-risk subtypes of human papillomavirus (HPV) is associated with the development of cervical cancer [39]. HPV infects epithelial cells, and, after integration in host DNA, the production of oncoproteins, mainly E6 and E7, disrupts natural tumor suppressor pathways and is required for proliferation of cervical carcinoma cells [40]. HPV also is believed to play a role in other human cancers, such as head and neck tumors, skin cancers in immunosuppressed patients, and other anogenital cancers.Cervical cancer is the second leading cause of cancer mortality in women worldwide, causing 240,000 deaths annually [41]. Of approximately 490,000 cases reported each year, more than 80 percent occur in the developing world, where effective but costly Pap smear screening programs are not in place [41]. Early precancerous changes and early cancers detected by Pap smears are effectively treated and cured with surgical therapy or ablation. In the absence of effective screening, the disease is detected late. Traditional therapeutic options for cervical cancer that have advanced beyond definitive surgical treatment are chemotherapy and radiation therapy, which are associated with many toxicities and do not offer a lasting cure.The immune system plays an important role in the prevention of persistent HPV infection and progression of precancerous lesions. Human papillomavirus is a poor natural immunogen; as a double stranded DNA virus, there is no RNA intermediate, nor does infection cause cytolysis, allowing initiation of innate immune responses [42]. HPV mainly encodes non-secreted nucleoproteins, which are poorly cross-presented and compared to other viruses its non-structural proteins are expressed at low levels. However, genital infection with HPV is usually transient. Additionally, inadequate T cell responses may lead to failure to clear HPV-infected cells. AIDS patients, renal transplant patients receiving immunosuppressive therapy, and individuals with T cell deficiencies have increased rates of HPV persistence, anogenital lesions, and cervical cancer [43-46].In 2006, an effective prophylactic vaccine against HPV 16 and 18 based on virus-like particles (VLP) of recombinant L1, the major capsid protein [47,48], was approved for use by the FDA based on clinical trials that demonstrated nearly 100 percent protection from persistent infection through the generation of high levels of neutralizing antibodies. Since these types are the causative agent of approximately 70 percent of cervical cancers, development of such an effective vaccine holds much promise for the prevention of cervical cancer [47]. However, the vaccine currently costs $360 for a complete course of three injections given over six months, does not provide protection against other high risk HPV types, will presumably have limited benefit to women already infected, and has an unknown duration of protection.Because of these limitations, therapeutic vaccination is being explored to treat women already infected and accelerate the impact of prophylactic vaccination in decreasing cervical cancer incidence. Traditional therapy for early cervical cancer and precancerous lesions involves surgical excision or ablation. Therapeutic vaccination seeks to generate a population of cytoxic T cells that will recognize and kill tumor cells. Since patients with T cell deficiencies are known to be more susceptible to HPV infection and disease progression, boosting T cell responses to HPV may be crucial to a therapeutic immune strategy. In the case of cervical cancer, E6 and E7 oncoproteins are expressed in all malignancies and are not found in uninfected normal cells. Therefore, they represent ideal targets for a therapeutic immune response. A number of strategies to generate immune responses against these antigens are under investigation. Viral and bacterial vectors have been used in mouse models to generate immune responses. Vaccinia virus delivery of HPV 16 and 18 modified E6 and E7 proteins has demonstrated safety and specific immune responses in early clinical trials [49]. DNA vaccination strategies also are under active investigation, and several are in various stages of clinical trials. Vaccination with plasmid DNA encapsulated in biodegradable micorparticles has shown histological and immunological responses when used to treat patients with high grade cervical dysplasia [50-52].Human T lymphotropic virus type I (HTLV-1)HTLV-1 is a slow transforming, single stranded RNA retrovirus and is associated with adult T-cell leukemia [53]. It possesses a diploid genome similar to other retroviruses: two long terminal repeats flanking gag, pol, and env genes as well as a number of accessory genes. HTLV-1 has a worldwide distribution, with an estimated 12 to 25 million people infected. However, disease is only observed in less than 5 percent of infected individuals. It is transmitted through blood transfusions, sexual contact, and during parturition. HTLV-1 displays a special tropism for CD4 cells, which clonally proliferate in adult T cell leukemia, though how this is effected is not known.HTLV-1 infection has a very long latency period of 20 to 30 years, but once tumor formation begins, progression is rapid. Standard chemotherapy often can bring about an initial response with a partial or complete remission; however, relapse is common, and median survival is eight months. The HTLV-1 Tax gene has been postulated to play an important role in tumorgenesis [54] through the activation of viral transcription and the hijacking of cellular growth and cell division machinery, but the mechanisms leading to adult T cell leukemia are not well understood. It has been suspected that HTLV-1 infection may not be sufficient to transform, and recent evidence suggests that the decreased diversity, frequency, and function of HTLV-1 specific CD8 T cells in the host may play an important part in the development of adult T-cell leukemia [55]. Therefore, targeted therapies using peptide, recombinant protein, DNA, and viral vectors with the goal of generating neutralizing antibody against HTLV-1 and multivalent cytotoxic T cell response against Tax are under investigation [56].SummaryThe viruses reviewed here illustrate the diverse biological pathways to malignancy and the challenges of treating the resulting diseases. Yet the presence of the viral gene products in cancer and precancerous cells present attractive targets that may be exploited in novel therapies that distinguish these cells from normal cells. Antivirals such as lamuvidine used in heptatitis B and ganciclovir for Kaposi sarcoma specifically target the viral replication machinery. Targeting cancer cells specifically would have advantages over traditional modalities such as chemotherapy and radiation, which can include significant toxicities. Cervical cancer, because it retains HPV viral oncoproteins E6 and E7 and requires their continued expression for proliferation, provides an ideal model for cytotoxic immune therapies against these known antigens.Given the prevalence of these cancers in the developing world and the limitations of health care infrastructure, strategies for vaccine design to prevent primary infection and targeted therapies for the treatment of disease must be carefully considered in this context. Use of needles, refrigeration, multiple doses, and cost are all significant barriers to the delivery of an effective vaccine [41]. Cost, need for trained personnel and sophisticated equipment and facilities may impede global use of the most advanced targeted therapies. These challenges suggest that exploration of prophylactic strategies and development of specific, targeted therapies are both necessary to decrease this portion of the global cancer burden.AcknowledgmentsI would like to thank Daniel DiMaio for his critical reading of this manuscript.AbbreviationsEBV Epstein-Barr virusHHV-8 human herpesvirusHPV human papillomavirusHTLV-1 human T lymphotropic virus type 1Article informationYale J Biol Med. 2006 Dec; 79(3-4): 115–122.Published online 2007 Oct.PMCID: PMC1994798Cancer IssueJohn B LiaoDepartment of Obstetrics, Gynecology, and Reproductive Services, Yale University School of Medicine, New Haven, ConnecticutTo whom all correspondence should be addressed: John B. Liao, Yale University School of Medicine, P.O. Box 208005, New Haven, CT 06520. Tel: 203-785-2685; Fax: 203-785-6765; E-mail: [email protected] B. Liao is a Berlex-NICHD Scholar of the Reproductive Scientist Development Program supported by NIH grant #5K12HD008949.Copyright ©2006, Yale Journal of Biology and MedicineThis is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License, which permits for noncommercial use, distribution, and reproduction in any digital medium, provided the original work is properly cited and is not altered in any way.This article has been cited by other articles in PMC.Articles from The Yale Journal of Biology and Medicine are provided here courtesy of Yale Journal of Biology and MedicineReferenceszur Hausen H. Viruses in human cancers. Science. 1991;254(5035):1167–1173. [PubMed]World Health Organization. Hepatitis C: global prevalence. Wkly Epidemiol Rec. 1997;72(46):341–344. [PubMed]Fattovich G, et al. Morbidity and mortality in compensated cirrhosis type C: a retrospective follow-up study of 384 patients. Gastroenterology. 1997;112(2):463–472. [PubMed]Beasley RP. Hepatitis B virus. The major etiology of hepatocellular carcinoma. Cancer. 1988;61(10):1942–1956. [PubMed]Lavanchy D. Hepatitis B virus epidemiology, disease burden, treatment, and current and emerging prevention and control measures. J Viral Hepat. 2004;11(2):97–107. [PubMed]The Cancer of the Liver Italian Program (CLIP) investigators. A new prognostic system for hepatocellular carcinoma: a retrospective study of 435 patients. Hepatology. 1998;28(3):751–755. [PubMed]Okuda K, et al. Natural history of hepatocellular carcinoma and prognosis in relation to treatment. Study of 850 patients. Cancer. 1985;56(4):918–928. [PubMed]Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet. 2003;362(9399):1907–1917. [PubMed]Ni YH, et al. Hepatitis B virus infection in children and adolescents in a hyperendemic area: 15 years after mass hepatitis B vaccination. Ann Intern Med. 2001;135(9):796–800. [PubMed]Ramsay M, et al. Control of hepatitis B in the United Kingdom. Vaccine. 1998;16(Suppl):S52–S55. [PubMed]Iwarson S. Report from Working Group 3 (the Czech Republic, Denmark, Finland, Norway, The Netherlands, Slovakia, Sweden and the UK) Vaccine. 1998;16(Suppl):S63–S64. [PubMed]Niederau C, et al. Long-term follow-up of HBeAg-positive patients treated with interferon alfa for chronic hepatitis B. N Engl J Med. 1996;334(22):1422–1427. [PubMed]Lampertico P, et al. Long-term suppression of hepatitis B e antigen-negative chronic hepatitis B by 24-month interferon therapy. Hepatology. 2003;37(4):756–763. [PubMed]Yuen MF, et al. Long-term follow-up of interferon alfa treatment in Chinese patients with chronic hepatitis B infection: The effect on hepatitis B e antigen seroconversion and the development of cirrhosis-related complications. Hepatology. 2001;34(1):139–145. [PubMed]Liaw YF, et al. Lamivudine for patients with chronic hepatitis B and advanced liver disease. N Engl J Med. 2004;351(15):1521–1531. [PubMed]Lai CL, et al. Prevalence and clinical correlates of YMDD variants during lamivudine therapy for patients with chronic hepatitis B. Clin Infect Dis. 2003;36(6):687–696. [PubMed]Manns MP, Wedemeyer H, Cornberg M. Treating viral hepatitis C: efficacy, side effects, and complications. Gut. 2006;55(9):1350–1359. [PMC free article] [PubMed]Schiff ER. Prevention of mortality from hepatitis B and hepatitis C. Lancet. 2006;368(9539):896–897. [PubMed]Terrault NA, Berenguer M. Treating hepatitis C infection in liver transplant recipients. Liver Transpl. 2006;12(8):1192–1204. [PubMed]Bartenschlager R. Hepatitis C virus replicons: potential role for drug development. Nat Rev Drug Discov. 2002;1(11):911–916. [PubMed]Mercer DF, et al. Hepatitis C virus replication in mice with chimeric human livers. Nat Med. 2001;7(8):927–933. [PubMed]Meuleman P, et al. Morphological and biochemical characterization of a human liver in a uPA-SCID mouse chimera. Hepatology. 2005;41(4):847–856. [PubMed]Borza CM, Hutt-Fletcher LM. Alternate replication in B cells and epithelial cells switches tropism of Epstein-Barr virus. Nat Med. 2002;8(6):594–599. [PubMed]Papadopoulos EB, et al. Infusions of donor leukocytes to treat Epstein-Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation. N Engl J Med. 1994;330(17):1185–1191. [PubMed]Haque T, et al. Treatment of Epstein-Barr-virus-positive post-transplantation lymphoproliferative disease with partly HLA-matched allogeneic cytotoxic T cells. Lancet. 2002;360(9331):436–442. [PubMed]Gottschalk S, et al. An Epstein-Barr virus deletion mutant associated with fatal lymphoproliferative disease unresponsive to therapy with virus-specific CTLs. Blood. 2001;97(4):835–843. [PubMed]Jackman WT, et al. Expression of Epstein-Barr virus gp350 as a single chain glycoprotein for an EBV subunit vaccine. Vaccine. 1999;17(7-8):660–668. [PubMed]Thorley-Lawson DA, Poodry CA. Identification and isolation of the main component (gp350-gp220) of Epstein-Barr virus responsible for generating neutralizing antibodies in vivo. J Virol. 1982;43(2):730–736. [PMC free article] [PubMed]Gu SY, et al. First EBV vaccine trial in humans using recombinant vaccinia virus expressing the major membrane antigen. Dev Biol Stand. 1995;84:171–177. [PubMed]Donati D, et al. Clearance of circulating Epstein-Barr virus DNA in children with acute malaria after antimalaria treatment. J Infect Dis. 2006;193(7):971–977. [PubMed]Ho JH. An epidemiologic and clinical study of nasopharyngeal carcinoma. Int J Radiat Oncol Biol Phys. 1978;4(3-4):182–198. [PubMed]Chang Y, et al. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma. Science. 1994;266(5192):1865–1869. [PubMed]Sarid R, Olsen SJ, Moore PS. Kaposi's sarcoma-associated herpesvirus: epidemiology, virology, and molecular biology. Adv Virus Res. 1999;52:139–232. [PubMed]Flore O, et al. Transformation of primary human endothelial cells by Kaposi's sarcoma-associated herpesvirus. Nature. 1998;394(6693):588–592. [PubMed]Martin DF, et al. Oral ganciclovir for patients with cytomegalovirus retinitis treated with a ganciclovir implant. Roche Ganciclovir Study Group. N Engl J Med. 1999;340(14):1063–1070. [PubMed]Bais C, et al. G-protein-coupled receptor of Kaposi's sarcoma-associated herpesvirus is a viral oncogene and angiogenesis activator. Nature. 1998;391(6662):86–89. [PubMed]Montaner S, et al. The Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor as a therapeutic target for the treatment of Kaposi's sarcoma. Cancer Res. 2006;66(1):168–174. [PubMed]Gingues S, Gill MJ. The impact of highly active antiretroviral therapy on the incidence and outcomes of AIDS-defining cancers in Southern Alberta. HIV Med. 2006;7(6):369–377. [PubMed]Wallin KL, et al. Type-specific persistence of human papillomavirus DNA before the development of invasive cervical cancer. N Engl J Med. 1999;341(22):1633–1638. [PubMed]von Knebel Doeberitz M, et al. Correlation of modified human papilloma virus early gene expression with altered growth properties in C4-1 cervical carcinoma cells. Cancer Res. 1988;48(13):3780–3786. [PubMed]Initiative for Vaccines Research Team. Geneva, Switzerland: World Health Organization; 2005. Jun, State of the art of vaccine research and development. 99 pp.Frazer IH. Prevention of cervical cancer through papillomavirus vaccination. Nat Rev Immunol. 2004;4(1):46–54. [PubMed]Palefsky J, Holly E. Immunosuppression and co-infection with HIV. J Natl Cancer Inst Monogr. 2003;31:41–46. [PubMed]Halpert R, et al. Human papillomavirus and lower genital neoplasia in renal transplant patients. Obstet Gynecol. 1986;68(2):251–258. [PubMed]Petry KU, et al. Cellular immunodeficiency enhances the progression of human papillomavirus-associated cervical lesions. Int J Cancer. 1994;57(6):836–840. [PubMed]Frisch M, Biggar RJ, Goedert JJ. Human papillomavirus-associated cancers in patients with human immunodeficiency virus infection and acquired immunodeficiency syndrome. J Natl Cancer Inst. 2000;92(18):1500–1510. [PubMed]Koutsky LA, et al. A controlled trial of a human papillomavirus type 16 vaccine. N Engl J Med. 2002;347(21):1645–1651. [PubMed]Harper DM, et al. Efficacy of a bivalent L1 virus-like particle vaccine in prevention of infection with human papillomavirus types 16 and 18 in young women: a randomised controlled trial. Lancet. 2004;364(9447):1757–1765. [PubMed]Kaufmann AM, et al. Safety and immunogenicity of TA-HPV, a recombinant vaccinia virus expressing modified human papillomavirus (HPV)-16 and HPV-18 E6 and E7 genes, in women with progressive cervical cancer. Clin Cancer Res. 2002;8(12):3676–3685. [PubMed]Garcia F, et al. ZYC101a for treatment of high-grade cervical intraepithelial neoplasia: a randomized controlled trial. Obstet Gynecol. 2004;103(2):317–326. [PubMed]Sheets EE, et al. Immunotherapy of human cervical high-grade cervical intraepithelial neoplasia with microparticle-delivered human papillomavirus 16 E7 plasmid DNA. Am J Obstet Gynecol. 2003;188(4):916–926. [PubMed]Klencke B, et al. Encapsulated plasmid DNA treatment for human papillomavirus 16-associated anal dysplasia: a Phase I study of ZYC101. Clin Cancer Res. 2002;8(5):1028–1037. [PubMed]Gallo RC, et al. Association of the human type C retrovirus with a subset of adult T-cell cancers. Cancer Res. 1983;43(8):3892–3899. [PubMed]Duggan DB, et al. HTLV-I-induced lymphoma mimicking Hodgkin's disease. Diagnosis by polymerase chain reaction amplification of specific HTLV-I sequences in tumor DNA. Blood. 1988;71(4):1027–1032. [PubMed]Kozako T, et al. Reduced frequency, diversity, and function of human T cell leukemia virus type 1-specific CD8+ T cell in adult T cell leukemia patients. J Immunol. 2006;177(8):5718–5726. [PubMed]Lynch MP, Kaumaya PT. Advances in HTLV-1 peptide vaccines and therapeutics. Curr Protein Pept Sci. 2006;7(2):137–145. [PubMed]

People Like Us

I love the way you can just insert a text box, or erase. Wow its so nice to be able to do this with documents.

Justin Miller